CN112368299A - Connexin 43 antibodies and uses thereof - Google Patents

Connexin 43 antibodies and uses thereof Download PDF

Info

Publication number
CN112368299A
CN112368299A CN201980024875.6A CN201980024875A CN112368299A CN 112368299 A CN112368299 A CN 112368299A CN 201980024875 A CN201980024875 A CN 201980024875A CN 112368299 A CN112368299 A CN 112368299A
Authority
CN
China
Prior art keywords
ser
antibody
val
thr
pro
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN201980024875.6A
Other languages
Chinese (zh)
Inventor
张彦丰
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Alamab Therapeutics Co ltd
Alamab Therapeutics Inc
Original Assignee
Alamab Therapeutics Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Alamab Therapeutics Co ltd filed Critical Alamab Therapeutics Co ltd
Priority to CN202310053523.1A priority Critical patent/CN116854816A/en
Publication of CN112368299A publication Critical patent/CN112368299A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Abstract

The present disclosure relates to compositions and methods for treating diseases or conditions associated with insufficient opening of Cx43 hemichannels in bone cells, preferably for treating cancer, cancer metastasis, osteosarcoma, osteoporosis, or osteopenia.

Description

Connexin 43 antibodies and uses thereof
Cross Reference to Related Applications
This application claims priority to U.S. provisional application No. 62/651,668, filed 2018, 4/2, the disclosure of which is incorporated herein by reference in its entirety.
Sequence listing
An ASCII text file named "172628 _010900_ sequence. txt" with a size of 43,787 bytes created by EFS-Web on day 4/1 2019 is incorporated herein by reference in its entirety.
Technical Field
The present disclosure relates generally to anti-connexin (Cx)43 antibodies and their use in treating diseases or conditions associated with Cx43 hemichannel opening in bone cells, for example.
Background
Despite numerous prophylactic and therapeutic measures, cancer remains a major cause of death worldwide. Between 2010 and 2020, new cancer cases in the united states are expected to increase by about 24% in men, over one million per year, and 21% in women, over 900,000 per year. The type of cancer that is expected to increase most in both men and women is melanoma; prostate, kidney, liver and bladder cancer in men; in women, lung, breast, uterine and thymus cancers are present. Cancer is the second most common cause of death in the united states, accounting for nearly 1 out of 4 cases. Many cancers are difficult or nearly impossible to treat with current methods. Many cancers escape current treatment regimens, develop resistance to treatment, or relapse after treatment.
Cancer metastasis occurs when cancer spreads from the body part in which it originally developed (e.g., breast or prostate) to other body parts (e.g., liver or bone) and a secondary tumor is established. Bone is the most common site of cancer metastasis. Cancers that metastasize to bone include, but are not limited to, breast cancer, prostate cancer, lung cancer, and skin cancer (e.g., melanoma). Bone metastases can be identified in up to 75% of patients with advanced breast and prostate cancer. Bone metastasis (met) is associated with many significant clinical and quality of life consequences, such as, but not limited to, painful, pathological fractures, spinal cord and nerve compression, bone marrow infiltration, and impaired movement. In many cases, the systemic presence of cancer can also make the cancer difficult to cure.
Normal bone is composed of three major cell types: bone forms osteoblasts, and bone resorbs osteoblasts and osteocytes. Osteocytes account for about 95% of skeletal cells, and maintain the bone remodeling process by coordinating osteolytic and osteogenic activities. When cancer cells invade the bone, many normal bone functions are affected. Cancer cells interact with the local microenvironment, promoting cancer cell survival through skeletal destruction and vascularization.
Bone cells express a hemichannel called the connexin (Cx)43 hemichannel. These osteocyte hemichannels are normally closed and can open upon exposure to mechanical stimuli, resulting in the release of various factors in the skeletal microenvironment. Factors released by hemichannel opening may mediate other processes that reduce cancer cell migration and bone metastasis. Alendronate (AD) is a potent and commonly used bisphosphate drug that has been shown to open Cx43 hemichannels in bone cells. Bisphosphonates are a class of drugs known to be useful in the treatment of many skeletal diseases, including bone metastases. Administration of bisphosphonates has been shown to be associated with a reduction in the incidence of bone metastasis and a reduction in mortality in patients with breast cancer. AD is associated with reduced tumor growth as well as reduced bone destruction and pain. AD inhibits osteoclastic activity, inducing the opening of Cx43 hemichannels in osteocytes. However, administration of AD is often associated with a number of serious side effects.
Thus, there is a need for effective methods of treating cancer metastasis, for example, by opening a Cx43 hemichannel.
Disclosure of Invention
Provided herein are compositions and methods for treating diseases or conditions associated with Cx43 hemichannel opening (e.g., inadequate or defective opening) in bone cells, preferably for treating cancer, cancer metastasis, osteosarcoma, osteoporosis, or osteopenia.
In one aspect, provided herein are anti-Cx 43 antibodies, or antigen-binding fragments thereof, comprising:
first, second and third triple Complementary Determining Region (CDR) sequences having SEQ ID NOs 1, 2 and 3, respectively; and
having the first, second and third light chain Complementarity Determining Region (CDR) sequences of SEQ ID NOS 4,5 and 6, respectively.
In some embodiments, the antibody or fragment thereof may have a heavy chain variable region having the amino acid sequence of SEQ ID NO. 7 and a light chain variable region having the amino acid sequence of SEQ ID NO. 8.
In another aspect, provided herein are anti-Cx 43 antibodies or antigen-binding fragments thereof, comprising a heavy chain having an amino acid sequence selected from SEQ ID NOs 9-17, and a light chain having an amino acid sequence of SEQ ID No. 18.
In another aspect, provided herein is an antibody that binds to an epitope located within amino acid sequence FLSRPTEKTI (SEQ ID NO: 19). In some embodiments, an epitope may comprise one or more amino acids selected from the group consisting of: f1, S3, R4, P5, T6, E7, K8, T9 and I10 of SEQ ID NO 19. In one embodiment, the epitope consists of F1, S3, R4, P5, T6, E7, K8, T9, and I10 of SEQ ID NO 19. In some embodiments, the epitope may include all 10 amino acids of SEQ ID NO 19. In some embodiments, an epitope may consist of all 10 amino acids of SEQ ID NO 19.
In another aspect, provided herein are isolated anti-Cx 43 antibodies or antigen-binding fragments thereof, wherein the antibodies or fragments thereof cross-compete with any of the antibodies or fragments thereof disclosed herein for binding to Cx 43. In some embodiments, the antibody or fragment thereof promotes opening of a Cx43 hemichannel in bone cells.
In another aspect, provided herein is a pharmaceutical composition for promoting Gx43 hemichannel opening in bone cells, preferably for treating cancer, cancer metastasis, osteosarcoma, osteoporosis, or osteopenia, comprising an antibody or fragment thereof disclosed herein and a pharmaceutically acceptable carrier.
Also provided herein is the use of an antibody or fragment thereof disclosed herein for the preparation of a medicament to promote opening of a Cx43 hemichannel in bone cells, preferably for the treatment of cancer, cancer metastasis, osteosarcoma, osteoporosis, or osteopenia.
Also provided herein is a method of promoting Cx43 hemichannel opening in bone cells, preferably for treating cancer, cancer metastasis, osteosarcoma, osteoporosis, or osteopenia, comprising contacting bone cells with an effective amount of an antibody or fragment thereof disclosed herein.
Also provided herein is a method of treating a disease or condition associated with insufficient opening of a Cx43 hemichannel in bone cells, preferably for treating cancer, cancer metastasis, osteosarcoma, osteoporosis, or osteopenia, comprising administering to a patient in need thereof a therapeutically effective amount of an antibody or fragment thereof disclosed herein.
Brief description of the drawings
FIG. 1: antibodies are screened against boxplots of the raw data.
FIG. 2: letter maps of the full replacement analysis of peptide FLSRPTEKTI were probed with antibodies under high stringency conditions. The base sequence is listed below the figure, with the sense signal of the baseline sequence at the red line. The substitution at a given position is plotted at the signal strength recorded for that substitution.
FIG. 3: letter maps of the full replacement analysis of peptide FLSRPTEKTI were probed with antibodies under high stringency conditions. The base sequence is listed below the figure, with the sense signal of the baseline sequence at the red line. The substitution at a given position is plotted at the signal strength recorded for that substitution.
Detailed Description
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the compositions and methods of the present disclosure.
Disclosed herein are compositions and methods related to anti-Cx 43 antibodies or antigen-binding fragments thereof. In some embodiments, the compositions disclosed herein exhibit superior activity, pharmaceutical utility (e.g., reduced toxicity), stability, and/or developability (e.g., reduced production costs) relative to those disclosed in PCT publication nos. WO2015/027120 and WO2017/147561 (incorporated herein by reference in their entirety). In some embodiments, the advantages are unexpected.
Definition of
For convenience, certain terms used in the specification, examples, and appended claims are collected here. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this document belongs.
The following terms and words used herein shall have the following meanings:
the articles "a" and "an" are used herein to mean one or more than one (i.e., at least one) of the grammatical object of the article. For example, "an element" means one element or more than one element.
The term "about" as used herein means an acceptable error within 20%, more preferably within 10% and most preferably within 5% of the indicated value.
An "anti-Cx 43 antibody" is an antibody that immunospecifically binds to Cx43 (e.g., its extracellular domain). The antibody may be an isolated antibody. Such binding to Cx shows a value of, for example, K of not more than 1. mu.M, not more than 100nM or not more than 50nMDThe value is obtained. K can be determined by any method known to those skilled in the artDSuch as surface proton resonance experiments or cell binding experiments. The anti-Cx 43 antibody can be a monoclonal antibody or an antigen-binding fragment thereof.
An "antibody" as used herein is a protein comprising a binding domain that binds to an epitope of interest. The term antibody includes monoclonal antibodies comprising immunoglobulin heavy and light chain molecules, single heavy chain variable domain antibodies, and variants and derivatives thereof, including chimeric variants of monoclonal and single heavy chain variable domain antibodies. The binding domain is substantially encoded by an immunoglobulin gene or immunoglobulin gene fragment, wherein the protein immunospecifically binds to an antigen. Recognized immunoglobulin genes include kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as a number of immunoglobulin variable region genes. Light chains are classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD, and IgE, respectively. For most vertebrate organisms, including humans and mice, typical immunoglobulin building blocks include tetramers comprising two pairs of identical polypeptide chains, each pair having one "light" (about 25kD) and one "heavy" chain (about 50-70 kD). "VL" and "VH" refer to these light and heavy chains, respectively. "CL"and" CH"refers to the constant regions of the light and heavy chains. Each strip VLAnd VHThe three β -loops above are responsible for binding to the antigen and are called "complementarity determining regions" or "CDRs". The "Fab" (antigen binding fragment) region includes one constant region and one variable region from each of the heavy and light chains of an antibodyI.e. VL,CL,VHAnd CH1。
Antibodies include intact immunoglobulins and antigen-binding fragments thereof. The term "antigen-binding fragment" refers to an antibody polypeptide fragment that binds to an antigen or competes for antigen binding (i.e., specific binding) with an intact antibody (i.e., the intact antibody from which it was derived). Antigen-binding fragments can be produced by recombinant or biochemical methods well known in the art. Exemplary antigen binding fragments include Fv, Fab, Fab ', (Fab')2CDRs, paratopes and single chain Fv antibodies (scFv), wherein VHAnd VLThe strands are joined together (directly or via a peptide linker) to form a continuous polypeptide.
Antibodies also include variants, chimeric antibodies, and humanized antibodies. The term "antibody variant" as used herein refers to an antibody having a single or multiple mutation in the heavy and/or light chain. In some embodiments, the mutation is present in the variable region. In some embodiments, the mutation is present in the constant region. "chimeric antibody" refers to an antibody in which a portion of each of the heavy and light chain amino acid sequences is homologous to the corresponding sequence of an antibody derived from a particular species or belonging to a particular family, but the remaining segment of the chain is homologous to the corresponding sequence of another species or family. Typically in these chimeric antibodies, the light and heavy chain variable regions mimic the variable regions of an antibody derived from a mammalian species, while the constant regions are homologous to sequences of an antibody derived from another species. One significant advantage of these chimeric forms is the ability to conveniently derive the variable regions from currently known sequences, for example using readily available hybridomas or B cells from non-human host organisms, while deriving the constant regions from, for example, human cell preparations. Although the variable region has the advantage of being easy to prepare and its specificity is not affected by its source, a constant region from a human will be more difficult to elicit an immune response when the antibody is injected into a human individual than a corresponding antibody whose constant region is from a non-human source. However, the definition is not limited to this specific example. A "humanized" antibody is a molecule that has an antigen binding site substantially derived from an immunoglobulin of a non-human species, with the remaining immunoglobulin structure of the molecule based on the structure and/or sequence of a human immunoglobulin. The antigen binding site may include the complete variable region fused to the constant region or the Complementarity Determining Regions (CDRs) of a suitable framework region grafted only into the variable region. The antigen binding site may be wild-type or modified with one or more amino acid substitutions, for example modified to more resemble a human immunoglobulin. Some forms of humanized antibodies retain all of the CDR sequences (e.g., humanized mouse antibodies that comprise all six CDRs from a mouse antibody). Other forms of humanized antibodies have one or more CDRs (one, two, three, four, five or six) that have been altered relative to the original antibody, also referred to as one or more CDRs "derived" from the one or more CDRs.
As described herein, antibody amino acid residues may be numbered according to the general numbering system of Kabat (Kabat et al (1991) Sequences of Immunological hot Proteins (Sequences of Proteins of Immunological Interest), fifth edition NIH, Public Health Service (Besseda, Maryland).
The term "binding" as used herein in the context of binding between an antibody and an epitope of Cx43 as a target refers to the process of non-covalent interaction between molecules. Preferably, the binding is specific. The specificity of an antibody can be determined based on affinity. Binding affinity or dissociation constant K of specific antibodies to their epitopesDLess than 10-7M, preferably less than 10-8M。
The term "affinity" refers to the strength of the binding reaction between the binding domain of an antibody and an epitope. It is the sum of the attractive and repulsive forces acting between the binding domain and the epitope. The term affinity as used herein refers to the dissociation constant KD
The term "antigen" refers to a molecule or portion of a molecule that binds to a selective binding molecule, such as an antibody, and can also be used in animals to produce antibodies that bind to an epitope of the antigen. An antigen may have one or more epitopes.
The term "cancer" broadly refers to uncontrolled, abnormal growth of the host's own cells, resulting in invasion of surrounding tissues and possibly tissues distal to the original site of abnormal cell growth in the host. The main category includes tumors, which are epidermal tissue cancers (e.g., skin, squamous cells); sarcomas, which are connective tissue carcinomas (e.g., bone, cartilage, fat, muscle, blood tissue, etc.); leukemia, which is a hematopoietic tissue cancer (e.g., bone marrow tissue); lymphomas and myelomas, which are immune cell carcinomas; and central nervous system cancers, including cancers of the brain and spinal cord tissue. "cancer," "neoplasm," and "tumor" are used interchangeably herein. As used herein, "cancer" refers to all types of cancer or neoplasms or malignancies, including leukemias, tumors, and sarcomas, whether new born or recurrent. Specific examples of cancer include: tumors, sarcomas, myelomas, leukemias, lymphomas, and mixed types of tumors. Non-limiting examples of cancers are new or recurrent cancers of the brain, myeloma, bladder, breast, cervix, head and neck, kidney, non-small cell lung, mesothelioma, ovary, prostate, sarcoma, stomach, uterus and medulloblastoma.
The term "epitope" includes any determinant region, preferably a polypeptide determinant region, capable of specifically binding to an immunoglobulin or T cell receptor. In certain embodiments, epitope determinants include groups of chemically active surface molecules (e.g., amino acids, sugar side chains, phosphoryl groups, or sulfonyl groups), while in certain embodiments, epitope determinants may have specific three-dimensional structural characteristics and/or specific charge characteristics. In one embodiment, the epitope is a region of an antigen bound by an antibody. In some embodiments, an antibody is said to bind specifically to an antigen when it preferentially recognizes its antigen of interest in a complex mixture of proteins and/or macromolecules. Methods of epitope mapping are well known in the art, such as X-ray co-crystallography identification, array-based oligopeptide scanning, site-directed mutagenesis, high-throughput mutagenesis mapping, and hydrogen-deuterium exchange. Epitopes can be formed from contiguous amino acids or noncontiguous amino acids juxtaposed due to the tertiary structure folding of the protein. Epitopes formed by contiguous amino acids are generally maintained after contact with denaturing solvents, while epitopes formed by tertiary folding are generally lost after treatment with denaturing solvents. Epitopes typically comprise at least 3, more typically at least 5 or about 8-10 amino acids in a unique spatial conformation.
The site on the antibody that binds to the epitope is called the "paratope," which typically includes the amino acid residues immediately adjacent to the epitope once bound. See Sela-Culang et al, Front Immunol.2013; 4:302.
"immunohistochemistry" or "IHC" refers to the process of detection in cells or tissue sections that enables the binding of antibodies that immunospecifically recognize an antigen of interest in biological tissues and their subsequent detection. An overview of IHC technology can be found, for example, in Ramos-Vara et al, Veterinary Pathology January 2014 Vol 51 (1),42-87, incorporated herein in its entirety by reference. To evaluate IHC results, different typing and semi-quantitative scoring systems were developed. See, e.g., Fedchenko et al, Diagnostic Pathology, 2014; 9:221, herein incorporated by reference in its entirety. An example is the H-score, which is obtained by adding the results of multiplying the percentage of cells with a staining intensity order value (from 0 for "no signal" to 3 scores for "strong signal") by 300 possible values.
"immunospecific" or "immunospecifically" (sometimes used interchangeably with "specifically") refers to an antibody that binds to one or more epitopes of a representation of interest via a domain encoded by an immunoglobulin gene or immunoglobulin gene fragment, but does not substantially recognize and bind to other molecules in a sample comprising a mixed population of antigen molecules. Typically, using, for example, real-time unlabeled biolayer interferometry techniques, e.g.
Figure BDA0002716162770000081
HTX biosensors, or using surface plasmon resonance, e.g. BIACORETMOr K binding immunospecifically to homologous antigen as determined by solution affinity ELISADValues were not greater than 50 nM. The use of these techniques is well known in the art.
The term "surface plasmon resonance" refers to an optical phenomenon whereby real-time biomolecular interactions can be analyzed by detecting changes in protein concentration in a biosensor matrix, for example using BIACORETMSystem (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.).
Biolayer interferometry is a label-free technique for measuring biomolecular interactions. It is an optical analysis technique that analyzes two interference patterns of surface reflected white light: a layer of immobilized protein at the biosensor tip and a layer of internal reference. Any change in the number of molecules bound to the biosensor tip results in a change in the interference pattern that can be measured in real time (Abdiche, Y.N., et al, Analytical Biochemistry, (2008),377(2), 209-217). In some embodiments, the binding characteristics of some anti-Cx 43 antibodies disclosed herein are evaluated with a "real-time biolayer interferometer based biosensor (Octet HTX technology)".
The terms "cross-competition," "cross-blocking," "cross-blocked," and "cross-blocking" are used interchangeably herein to refer to the interference of an antibody or fragment thereof with the binding of a target Cx43, either directly or indirectly, through allosteric modulation of an anti-Cx 43 antibody herein. The extent to which an antibody or fragment thereof can interfere with the binding of another antibody or fragment thereof to a target and therefore whether cross-blocking or cross-competition can be determined in a competitive binding assay, which can be referred to in accordance with the present disclosure. One particularly suitable quantitative cross-competition assay uses FACS or AlphaScreen-based methods to determine the ability of a labeled (e.g., His-tag, biotinylated, or radiolabeled) antibody or fragment thereof to compete with another antibody or fragment thereof for binding to its target. In general, a cross-competitive antibody or fragment thereof is thus an antibody which is capable of binding to a target, for example in a cross-competition experiment, and therefore the displacement (displacement) of an immunoglobulin single variable region or polypeptide recorded according to the disclosure during the course of the experiment or in the presence of a second antibody or fragment thereof up to 100% of the maximum theoretical displacement (e.g. by cold (unlabeled) antibody or fragment thereof which is required to be cross-blocked) is displaced by a given amount of potential cross-blocking antibody or fragment thereof to be tested, for example in a FACS-based competition experiment. Preferably, therefore, the cross-competitive antibody or fragment thereof has between 10% and 100%, more preferably between 50% and 100% of the substitutions recorded.
Cross-competition between antibodies can also be determined using real-time label-free biolayer interferometry experiments. Cross-competition between the two antibodies can be expressed as binding of the second antibody is lower than the beijing signal due to self-binding (the first and second antibodies are the same antibody). Cross-competition between two antibodies can be expressed, for example, as% binding of the second antibody less than baseline self-background binding (the first and second antibodies are the same antibody).
The terms "promote", "enhance" and "induce" are used interchangeably herein to refer to any statistically significant increase in biological activity (e.g., hemichannel opening). For example, "promoting" may refer to an increase in biological activity of about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100%.
The term "patient" includes humans or other mammals that receive prophylactic or therapeutic treatment.
The term "treatment" as used herein refers to a therapeutic or prophylactic measure such as described herein. A "treatment" method employs administration of a Cx43 ligand provided herein to a patient (e.g., a patient having cancer) to prevent, cure, delay, reduce the severity of, or alleviate one or more symptoms of the cancer or recurrent cancer, or to extend the survival of the patient beyond that which would be expected in the absence of such treatment. The methods of "treating" also employ administering a Cx43 ligand (e.g., an antibody) provided herein to the patient to provide a cancer treatment in the patient that is beyond that which would be expected in the absence of such treatment.
As used herein, the term "effective amount" refers to an amount of an agent (e.g., a Cx43 ligand, such as an anti-Cx 43 antibody) that is sufficient to effect treatment, prognosis, or diagnosis of a cancer when administered to a patient. The therapeutically effective amount may vary depending on the patient and the disease or condition being treated, the weight and age of the subject, the severity of the disease or condition, and the mode of administration, etc., which can be readily determined by one of ordinary skill in the art. The dosage administered may range, for example, from about 1ng to about 10,000mg, from about 5ng to about 9,500mg, from about 10ng to about 9,000mg, from about 20ng to about 8,500mg, from about 30ng to about 7,500mg, from about 40ng to about 7,000mg, from about 50ng to about 6,500mg, from about 100ng to about 6,000mg, from about 200ng to about 5,500mg, from about 300ng to about 5,000mg, from about 400ng to about 4,500mg, from about 500ng to about 4,000mg, from about 1 μ g to about 3,500mg, from about 5 μ g to about 3,000mg, from about 10 μ g to about 2,600mg, from about 20 μ g to about 2,575mg, from about 30 μ g to about 2,550mg, from about 40 μ g to about 2,500mg, from about 50 μ g to about 2,475mg, from about 100 μ g to about 2,450mg, from about 200 μ g to about 2,85mg, from about 30 μ g to about 2,550mg, from about 40 μ g to about 2,500mg, from about 50 μ g to about 2,475mg, from about 1,500 mg to about 1,000mg, from about 1,84 mg to about 1,500 mg, from about 1,500 mg, about 3.0mg to about 975mg, about 3.5mg to about 950mg, about 4.0mg to about 925mg, about 4.5mg to about 900mg, about 5mg to about 875mg, about 10mg to about 850mg, about 20mg to about 825mg, about 30mg to about 800mg, about 40mg to about 775mg, about 50mg to about 750mg, about 100mg to about 725mg, about 200mg to about 700mg, about 300mg to about 675mg, about 400mg to about 650mg, about 500mg, or about 525mg to about 625mg of the antibody or antigen-binding fragment thereof provided herein. Administration may be, for example, weekly, biweekly, every three weeks, every 4 weeks, every 5 weeks, or every 6 weeks. The dosage regimen may be adjusted to provide the best therapeutic response. An effective amount is also one in which any toxic or deleterious (side effects) effects of the agent are minimized and/or offset by a beneficial effect. Administration can be intravenous, either exactly or about 6mg/kg or 12mg/kg weekly, or 12mg/kg or 24mg/kg biweekly. Other dosing regimens are described below.
One of ordinary skill in the appropriate art will be able to generally understand recombinant nucleic acid techniques, microbiology, immunology, antibody engineering, and other terms within the fields of molecular and cell biology as used herein. For example, recombinant DNA may be prepared, oligonucleotide synthesis may be performed, and tissue culture and transformation may be performed using conventional techniques (e.g., electroporation, transfection, or lipofection). Enzymatic reactions and purification techniques can be performed according to the manufacturer's instructions or according to routine procedures in the art or as described herein. The techniques and methods described above can be generally performed as described in numerous comprehensive and monographic documents as known in the art, and also as cited and discussed in this specification. See, e.g., Sambrook et al, 2001, Molecular Cloning: a Laboratory Manual (molecular cloning: A Laboratory Manual) (3 rd edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.), which is incorporated by reference herein for all purposes in its entirety. Unless otherwise indicated, nomenclature used in analytical chemistry, synthetic organic chemistry, and medical and pharmaceutical chemistry, and laboratory methods and techniques described herein are those well known and commonly used in the art. For chemical synthesis, chemical analysis, pharmaceutical preparation, formulation and delivery, and treatment of patients, standard techniques can be used.
As used herein, the terms "comprising" or "comprises" refer to compositions, methods, and components thereof that are present in a given embodiment, but do not contain the specified elements.
As used herein, the term "consisting essentially of … …" refers to those elements required for a given implementation. The terms allow for the presence of additional elements that do not materially affect the basic and novel or functional characteristics of this embodiment of the disclosure.
The term "consisting of … …" refers to the compositions, methods, and respective components thereof described herein, excluding any elements not listed in the description of the embodiments.
In this specification and the appended claims, the singular forms "a", "an", and "the" include plural referents unless the content clearly dictates otherwise. Thus, for example, "a method" includes one or more methods, and/or steps of the type described herein, which will become apparent to those skilled in the art upon reading this disclosure and so forth.
Various aspects and embodiments of the disclosure are described in further detail in the following subsections.
Cx43
The various cells are able to communicate with each other and with the extracellular environment through the gap junctions formed by the hemichannels and connexins. Connexins are ubiquitously expressed in humans. Six connexins constitute one half-channel, and two half-channels constitute one gap junction channel. Gap junctions are a group of channels located within the interstitial membranes of adjacent cells that mediate intercellular communication. A half channel is a separate entity from a gap connecting channel. Hemichannels allow molecular exchange between intracellular compartments and the extracellular environment.
Bone cells express a hemichannel called the connexin (Cx)43 hemichannel. These osteocyte hemichannels are normally closed and can open upon exposure to mechanical stimuli, resulting in the release of various factors in the skeletal microenvironment. Factors released by hemichannel opening may mediate other processes that reduce cancer cell migration and bone metastasis.
Connexin-43, also known as gap junction alpha-1 protein (GJA1), is a 4382kDa protein consisting of 382 amino acids (NCBI reference: NP-000156.1). GJA1 contains a long C-terminal tail, an N-terminal domain and multiple transmembrane domains. The protein passes four times through the phospholipid bilayer, leaving its C and N termini in contact with the cytoplasm. The C-terminal tail consists of 50 amino acids, including post-translational modification sites and binding sites for transcription factors, cytoskeletal elements and other proteins. As a result, the C-terminal tail is critical for regulating functions such as pH gating and channel assembly. Notably, the DNA region of the GJA1 gene (NCBI gene ID: 2697) encoding this tail is highly conserved, suggesting that it is either resistant to mutation or becomes lethal after mutation. At the same time, the N-terminal domain participates in channel gating and oligomerization, thus controlling the switching between the open and closed states of the channel. The transmembrane domains form gap junction channels, while the extracellular loops facilitate proper channel docking. In addition, the two extracellular loops form disulfide bonds that interact with the two hexamers, forming an intact gap junction channel.
anti-Cx 43 antibodies
Promoting or enhancing the opening of a Cx43 hemichannel can induce or promote the opening of a Cx43 hemichannel in bone cells, thereby treating, for example, cancer metastasis, osteosarcoma, osteoporosis, or osteopenia. Thus, anti-Cx 43 antibodies can be used as effective agents in the treatment of cancer.
In certain embodiments, the anti-Cx 43 antibody can be a monoclonal antibody or antigen-binding fragment thereof. In certain embodiments, the anti-Cx 43 antibody can be modified, e.g., chimeric or humanized antibodies derived from a mouse anti-Cx 43 antibody. In some embodiments, the anti-Cx 43 antibody is an antibody or antigen-binding fragment thereof that binds to an epitope present on a human Cx43 protein (e.g., extracellular loop or portion thereof).
An exemplary anti-Cx 43 antibody can have one or more of the following CDR sequences:
heavy chain:
CDR1(SEQ ID NO.:1):GYTFTSYY
CDR2(SEQ ID NO.:2):INPSNAGT
CDR3(SEQ ID NO.:3):TREGNPYYTMNY
light chain:
CDR1(SEQ ID NO.:4):QSLLESDGKTY
CDR2(SEQ ID NO.:5):LVS
CDR3(SEQ ID NO.:6):WQGTHFPWT
in some embodiments, it has been surprisingly found that antibodies having the CDR sequences described above exhibit superior binding affinity and/or antibody stability compared to antibodies disclosed in PCT publications WO2015/027120 and WO 2017/147561. Without wishing to be bound by theory, it is believed that the "NG" to "NA" mutation in heavy chain CDR2 reduces deamidation. Deamidation of antibodies, particularly in the CDR regions, can result in changes in binding affinity, degradation of the antibody, and changes in charge variants, thereby affecting antibody function and increasing antibody production costs. Thus, the CDRs disclosed herein provide improved binding affinity and antibody stability, resulting in advantageous technical effects over those disclosed in PCT publications WO2015/027120 and WO 2017/147561.
Monoclonal antibodies can be humanized and optimized using, for example, CDR grafting, germline modeling and 3-D structural analysis to increase the pharmaceutical usefulness and/or developability of the antibody. In some embodiments, a humanized anti-Cx 43 antibody can have one or both of the following variable domains:
heavy chain variable region (SEQ ID NO: 7):
EVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMYWVRQAPGQGLEWIGGINPSNAGTNFNEKF
KNRATLTVDKSTSTAYMELSSLRSEDTAVYYCTREGNPYYTMNYWGQGTLVTVSS
light chain variable region (SEQ ID NO: 8):
DVVMTQSPLSLPVTIGQPASISCKSSQSLLESDGKTYLNWLQQRPGQSPRRLIYLVSKLDSGVPDRFS
GSGSGTDFTLKISRVEAEDVGVYYCWQGTHFPWTFGGGTKVEIK
in selected embodiments, the anti-Cx 43 antibody may have a variable domain fused to a constant region of, for example, human IgG1 or IgG4, which may optionally comprise one or more mutations. In some embodiments, mutations can be designed to reduce or minimize the cytotoxic effector function of an antibody while maintaining binding affinity and antibody stability. For example, an anti-Cx 43 antibody can have one or more of the following heavy chain sequences (where the bold portions correspond to the variable regions and the non-bold portions correspond to the constant regions):
heavy chain of Ab # D (SEQ ID NO. 9)
Figure BDA0002716162770000131
Heavy chain of Ab # F (SEQ ID NO: 10)
Figure BDA0002716162770000132
Heavy chain of Ab # H (SEQ ID NO: 11)
Figure BDA0002716162770000133
Ab # J heavy chain (SEQ ID NO. 12)
Figure BDA0002716162770000141
Ab # L heavy chain (SEQ ID NO. 13)
Figure BDA0002716162770000142
Heavy chain of Ab # N (SEQ ID NO. 14)
Figure BDA0002716162770000143
Heavy chain of Ab # P (SEQ ID NO. 15)
Figure BDA0002716162770000144
Heavy chain of Ab # R (SEQ ID NO: 16)
Figure BDA0002716162770000145
Figure BDA0002716162770000151
Ab # T heavy chain (SEQ ID NO. 17)
Figure BDA0002716162770000152
In some embodiments, an anti-Cx 43 antibody can have one or more of the following light chain sequences (where the bold portions correspond to the variable regions and the non-bold portions correspond to the constant regions):
ab # F, Ab # H, Ab # J, Ab # L, Ab # N, Ab # P, Ab # R, light chain of Ab # T (SEQ ID NO: 18)
Figure BDA0002716162770000153
In yet another embodiment, the anti-Cx 43 antibody can comprise a mixture or blend of two or more anti-Cx 43 antibodies (cocktails), each anti-Cx 43 antibody binding to the same or a different epitope on Cx 43.
In some embodiments, bispecific antibodies can be prepared in which at least one specificity is an anti-Cx 43 antibody or antigen-binding fragment thereof disclosed herein. The other specificity may be for another target associated with the disease being treated.
In one aspect, there is provided use of a Cx43 ligand in the manufacture of a medicament. In another aspect, a method of inhibiting tumor growth and/or metastasis in a patient is provided, the method comprising administering to the patient an effective amount of a Cx43 ligand.
Preparation of anti Cx43 antibodies
anti-Cx 43 antibodies can be prepared using various methods commonly known in the art. For example, phage display technology can be used to screen human antibody libraries to generate fully human monoclonal antibodies for treatment. High affinity binders may be considered candidates for neutralization studies. In addition, conventional monoclonal methods can be used, wherein mice or rabbits can be immunized with human proteins, identified and tested candidate binders, and humanized antibodies ultimately produced by grafting the binding sites of the heavy and light chains into human antibody coding sequences.
Antibodies typically comprise two pairs of identical polypeptide chains, each pair having one full length "light" chain (typically having a molecular weight of about 25 kDa) and one full length "heavy" chain (typically having a molecular weight of about 50-70 kDa). The amino-terminal portion of each chain comprises a variable region of about 100-110 or more amino acids, typically responsible for antigen recognition. The carboxy-terminal portion of each chain is defined as the constant region that is generally responsible for effector function. The variable regions of each of the heavy and light chains typically exhibit the same overall structure, including four relatively conserved Framework Regions (FRs) connected by three hypervariable regions (also known as complementarity determining regions or CDRs). The CDRs of each pair of two chains are typically aligned by framework regions, and are capable of binding to a particular epitope. From N-terminus to C-terminus, both light and heavy chains comprise domains FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR 4. The amino acid assignments for each domain are generally associated with Kabat, Sequences of Proteins of Immunological Interest (sequence of immunothermomech protein) (National Institutes of Health (1987 and 1991) of Besserda, Maryland or Chothia and Lesk, J.mol.biol.196:901-917 (1987); chothia et al, Nature 342:878-883(1989) are in agreement.
With the development of monoclonal antibodies, antibodies have become useful and interesting drugs as drugs. Monoclonal antibodies can be produced using any method that produces antibody molecules by continuous cell lines in culture. Examples of suitable methods for preparing monoclonal antibodies include the hybridoma method of Kohler et al (1975, Nature 256: 495-497) and the human B-cell hybridoma method (Kozbor, 1984, J.Immunol.133: 3001; and Brodeur et al, 1987, monoclonal antibody production techniques and applications, Marcel Dekker, Inc, New York, pp 51-63).
Monoclonal antibodies can be modified for use as therapeutic agents. One example is a "chimeric" antibody in which a portion of the heavy and/or light chain is identical to or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain is identical to or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass. Other examples are fragments of such antibodies, as long as they exhibit the desired biological activity. See U.S. Pat. No. 4,816,567 and Morrison et al (1985), Proc. Natl. Acad. Sci. USA 81: 6851-6855. A related development is a "CDR-grafted" antibody, wherein the antibody comprises one or more Complementarity Determining Regions (CDRs) from a particular species or belonging to a particular antibody class or subclass, while the remainder of the antibody chain is identical or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass.
Another development is "humanized" antibodies. Methods for humanizing non-humanized antibodies are well known in the art (see U.S. Pat. Nos. 5,585,089 and 5,693,762; see also CecileVincke et al, J.biol. chem.2009; 284: 3273. and 3284 to humanize lambda antibodies). Typically, humanized antibodies are produced by non-human animals, and then certain amino acid residues, typically from the non-antigen-recognizing portion of the antibody, are modified to be homologous to those in a human antibody of the corresponding isotype. Humanization can be carried out, for example, using methods described in the art (Jones et al, 1986, Nature 321: 522-525; Riechmann et al, 1988, Nature 332: 323-327; Verhoeyen et al, 1988; (Science 239: 1534-1536), by replacing at least a portion of the rodent variable region with the corresponding region of a human antibody.
A recent development is human antibodies that do not contact human antigens ("fully human antibodies"). Such antibodies are produced by immunization with an antigen (typically having at least 6 contiguous amino acids), which may optionally be conjugated to a carrier, using a transgenic animal (e.g., a mouse) capable of producing a human antibody repertoire without producing endogenous mouse immunoglobulin. See, e.g., Jakobovits et al, 1993, Proc. Natl. Acad. Sci. USA 90: 2551-; jakobovits et al, 1993, Nature 362: 255-258; and Bruggermann et al, 1993, Yeast in Immunol.7: 33. In one example of these methods, transgenic animals are produced by incapacitating endogenous mouse immunoglobulin loci in which the mouse heavy and light chain immunoglobulin chains are encoded, and inserting the loci encoding human heavy and light chain proteins into their genomes. Partially modified animals with less than all of the modifications are then crossed to obtain animals with all of the desired immune system modifications. After administration of the immunogen, antibodies produced by these transgenic animals are immunospecific for these antigens having human (rather than murine) amino acid sequences, including the variable regions. See PCT publication Nos. WO96/33735 and WO94/02602, incorporated herein by reference. Other methods are described in U.S. Pat. No. 5,545,807, PCT publication Nos. WO91/10741, WO90/04036, and EP 546073B1 and EP 546073A1, which are incorporated by reference. Human antibodies can also be produced by expression of recombinant DNA in a host cell or in a hybridoma cell, as described herein.
In some embodiments, phage display technology can be used to screen for therapeutic antibodies. In phage display, antibody libraries can be displayed on the surface of filamentous phage, and the constructed libraries can be screened against phage that bind to an immunogen. Antibody phage is based on genetic engineering of phage as well as antigen-directed selection and iterative experiments of phage propagation. This technique allows the in vitro selection of Cx43 monoclonal antibodies. The phage display process begins with the preparation of an antibody library, followed by ligation of Variable Heavy (VH) and Variable Light (VL) PCR products into a phage display vector, and final analysis of monoclonal antibody clones. The VH and VL PCR products representing the antibody repertoire are ligated into a phage display vector (e.g., phagemid pComb3X) engineered to express VH and VL as scfvs fused to the pIII minor capsid proteins of filamentous phage of e.coli originally derived from the M13 phage. However, the phage display vector pComb3X does not have all the other genes necessary to encode a complete phage in E.coli. For those genes, helper phages were added to E.coli transformed with the phage display vector library. As a result, a library of phages, each expressing Cx43 monoclonal antibody on its surface and carrying a vector of the respective nucleotide sequence, was obtained. Phage display can also be used to produce the Cx43 monoclonal antibody itself (not linked to the phage coat protein) in certain strains of e. Following the VL and VH sequences, additional cdnas were engineered in phage display vectors to characterize and purify the produced mabs. In particular, the recombinant antibody may have a Hemagglutinin (HA) epitope tag and polyhistidine to facilitate purification from solution.
Multiple antibody phage libraries consisting of infection with about 10 of helper phage8Independent E.coli transformants were generated. Using biopanning, libraries that bind to the above-described immunogenic sequences or fragments thereof phage can be screened through the surface expressed by monoclonal antibodies. The cycle panning allowed the extraction of potentially very rare antigen-binding clones and consisted of multiple rounds of phage binding to antigen (immobilized on ELISA plates or in solution on the cell surface), washing, elution and re-amplification of phage binders in e. In each round, specific binders were selected from the pool by washing away unbound binders and selectively eluting bound phage clones. After three or four rounds, phage clones were characterized by their surface Cx43 monoclonal antibody binding with high specificity for targeted selection on immobilized immunogens.
Another method is to add a C-terminal His tag suitable for purification by affinity chromatography to the immunogenic sequences listed above. The purified protein can be inoculated into mice with a suitable adjuvant. Monoclonal antibodies produced in the hybridomas can be tested for binding to the immunogen, and positive binders can be screened as described in the assays herein.
Fully human antibodies can also be generated from phage display libraries (as disclosed in Hoogenboom et al, 1991, J.mol.biol.227: 381 and Marks et al, 1991, J.mol.biol.222: 581). These processes mimic immunoselection by displaying antibody libraries on the surface of filamentous phage and subsequently selecting by binding to selected antigens. One such technique is described in PCT publication No. WO99/10494, which is incorporated herein by reference, which describes the use of this method to isolate high affinity and functional agonistic antibodies for MPL-and msk-receptor antibodies.
The nucleotide sequence encoding the above antibody can be determined. Thereafter, chimeric, CDR grafted, humanized and fully human antibodies can also be produced by recombinant methods. Nucleic acids encoding the antibodies can be introduced into host cells and expressed using materials and methods generally known in the art.
Provided herein are antibodies directed to Cx 43. Preferably the antibody binds Cx 43. In preferred embodiments, the present invention provides nucleotide sequences encoding heavy and light chain immunoglobulin molecules and amino acid sequences comprising the same, particularly sequences corresponding to the variable regions thereof. In preferred embodiments, sequences corresponding to the CDRs, particularly from CDR1 to CDR3, are provided. In further embodiments, the disclosure provides hybridoma cell lines expressing such immunoglobulin molecules and monoclonal antibodies produced therefrom, preferably purified human monoclonal antibodies directed to human Cx 43.
The CDRs of the light and heavy chain variable regions of the anti-Cx 43 antibodies of the present disclosure may be grafted to Framework Regions (FRs) from the same or another species. In certain embodiments, the CDRs of the light and heavy chain variable regions of the anti-Cx 43 antibody can be grafted to consensus human FRs. To generate consensus human FRs, FRs from several human heavy or light chain amino acid sequences are aligned to identify a consensus amino acid sequence. The FRs of the anti-Cx 43 antibody heavy or light chain may be replaced by FRs from a different heavy or light chain. Rare amino acids in the anti-Cx 43 antibody heavy and light chain FRs are typically not substituted, while the remaining FR amino acids may be substituted. Rare amino acids are specific amino acids, which are located at positions not normally present in the FR. Grafted variable regions of anti-Cx 43 antibodies from the present disclosure can be used with constant regions that are different from the constant regions of anti-Cx 43 antibodies. Alternatively, the grafted variable region is part of a single chain Fv antibody. CDR grafting is described, for example, in U.S. patent nos. 6,180,370, 5,693,762, 5,693,761, 5,585,089, and 5,530,101, which are incorporated herein by reference for any purpose.
In some embodiments, the antibodies of the invention can be produced by a hybridoma cell line. In these embodiments, the antibodies of the invention bind to Cx43 with a dissociation constant (KD) of between about 4pM and 1 μ M. In certain embodiments of the disclosure, the antibody binds Cx43 with a KD of less than about 100nM, less than about 50nM, or less than about 10 nM.
In embodiments, the antibody of the present disclosure is an IgG1, IgG2, IgG3, or IgG4 isotype, e.g., IgG1 isotype. In certain embodiments, the antibody comprises a human kappa or lambda light chain and a human IgG1, IgG2, or IgG4 heavy chain. In embodiments, the variable region of the antibody is linked to a constant region of an IgG1, IgG2, or IgG4 isotype. In embodiments, the variable region of the antibody is linked to a constant region other than that of the IgG1, IgG2, or IgG4 isotype. In certain embodiments, the antibodies of the present disclosure have been cloned for expression in mammalian cells.
In alternative embodiments, the antibodies of the invention may be expressed in cell lines other than hybridoma cell lines. In these embodiments, sequences encoding particular antibodies may be used to transform a suitable mammalian host cell. According to these embodiments, transformation can be accomplished using any known method for introducing a polynucleotide into a host cell, including, for example, packaging the polynucleotide in a virus (or viral vector) and transducing the host cell with the virus (or vector) or by transfection methods known in the art. Such methods are described, for example, in U.S. Pat. nos. 4,399,216, 4,912,040, 4,740,461, and 4,959,455 (which are all incorporated herein by reference for any purpose). In general, the transformation method used may depend on the host to be transformed. Methods for introducing heterologous polynucleotides into mammalian cells are well known in the art and include, but are not limited to, dextran-mediated transfection, calcium phosphate precipitation, polyene-mediated transfection, protoplast fusion, electroporation, encapsulation of polynucleotides in liposomes, and direct microinjection of DNA into the nucleus of a cell.
According to certain embodiments of the methods of the present disclosure, a nucleic acid molecule of the present disclosure encoding an amino acid sequence of a heavy chain constant region, a heavy chain variable region, a light chain constant region, or a light chain variable region of a Cx43 antibody is inserted into a suitable expression vector using standard ligation techniques. In a preferred embodiment, the Cx43 antibody heavy or light chain constant region is appended to the C-terminus of the appropriate variable region and ligated into an expression vector. The vector is typically selected to function in the particular host cell being used (i.e., the vector is compatible with the host cell machinery such that amplification of the gene and/or expression of the gene may occur). For a review of expression vectors, see Goeddel eds.), 1990, meth.enzymol.vol.185, Academic press.n.y.
In general, expression vectors used in any host cell may contain sequences for plasmid maintenance as well as for cloning and expression of exogenous nucleotide sequences. Such sequences typically include one or more of the following nucleotide sequences: a promoter, one or more enhancer sequences, an origin of replication, a transcription termination sequence, complete intron sequences containing donor and acceptor splice sites, sequences encoding polypeptide secretion leader sequences, ribosome binding sites, polyadenylation sequences, a polylinker region for insertion of a nucleic acid encoding a polypeptide to be expressed, and selectable marker elements. These sequences are well known in the art.
The expression vectors of the present disclosure can be constructed from starting vectors, such as commercially available vectors. Such vectors may or may not contain all of the desired flanking sequences. When one or more of the flanking sequences described above are not already present in the vector, they may be obtained separately and ligated into the vector. Methods for obtaining each flanking sequence are well known to those skilled in the art.
After constructing the vector and inserting the nucleic acid molecule encoding the light chain or the heavy chain or the light and heavy chains comprising the anti-Cx 43 antibody into the appropriate site of the vector, the complete vector can be inserted into a suitable host for amplifying and/or polypeptide expressing cells. Transformation of the expression vector for the anti-Cx 43 antibody into a selected host cell can be accomplished by well-known methods, including transfection, infection, calcium phosphate co-precipitation, electroporation, microinjection, lipofection, DEAE-dextran mediated transfection or other known techniques. The method of choice will depend in part on the type of host cell used. These and other suitable methods are well known to the skilled person and are set out, for example, in Sambrook et al, supra.
When the host cells are cultured under appropriate conditions, the anti-Cx 43 antibody is synthesized and can then be collected from the culture medium (if the host cells secrete it into the culture medium) or directly from the host cells producing it (if it is not secreted). The choice of a suitable host cell will depend on a variety of factors, such as the desired expression level, the modification of the polypeptide that is desirable or necessary for activity (e.g., glycosylation or phosphorylation), and the ease with which it folds into a biologically active molecule.
Mammalian cell lines useful as expression hosts are well known in the art and include, but are not limited to, a number of immortalized cell lines available from the American Type Culture Collection (ATCC), including, but not limited to, Chinese Hamster Ovary (CHO) cells, Human Embryonic Kidney (HEK), HeLa cells, hamster kidney (BHK) cells, monkey kidney Cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), and a number of other cell lines. In certain embodiments, cell lines can be selected by determining which cell lines have high expression levels and produce antibodies with constitutive Cx43 binding properties. In another embodiment, cell lines that do not produce their autoantibodies but have the ability to produce and secrete heterologous antibodies can be selected from the B cell lineage (e.g., mouse myeloma cell lines NS0 and SP 2/0).
Epitope mapping and related techniques
The present disclosure provides anti-Cx 43 antibodies that interact with one or more amino acids found within one or more domains, such as extracellular loops, of the Cx43 molecule. The epitope bound by the antibody may comprise a contiguous sequence of 2 or more (e.g., 2, 3, 4,5, 6,7, 8, 9 or more) amino acids located within one or more extracellular loops. Alternatively or additionally, an epitope may comprise 1 or more non-contiguous amino acids (or amino acid sequence) located within one or more extracellular loops (e.g., conformational epitopes).
Various techniques known to those of ordinary skill in the art can be used to determine whether an antibody "interacts with one or more amino acids within a polypeptide or protein. Exemplary techniques include, for example, conventional cross-blocking experiments such as those described in Antibodies, Harlow and Lane (cold spring harbor press, cold spring harbor, new york). Other Methods include alanine scanning mutation analysis, peptide blot analysis (Reineke (2004) Methods mol. biol.248: 443-63), peptide cleavage analysis crystallography studies, and NMR analysis. In addition, methods such as epitope excision, epitope extraction and chemical modification of antigen can be employed (Tomer (2000) prot. Sci.9: 487-496).
Another method that can be used to identify amino acids within a polypeptide for antibody interactions is to detect hydrogen/deuterium exchange by mass spectrometry. Generally, the hydrogen/deuterium exchange method involves deuterium labeling the target protein, and then binding the antibody to the deuterium labeled protein. Next, the protein/antibody complex is transferred into water, and the exchangeable protons within the amino acids protected by the antibody complex undergo reverse exchange from deuterium to hydrogen at a slower rate than the exchangeable protons within the amino acids not belonging to the interface. As a result, amino acids forming part of the protein/antibody interface may retain deuterium and therefore have a relatively high mass compared to amino acids not included in the interface. After antibody dissociation, the target protein is subjected to protease cleavage and mass spectrometry analysis, revealing deuterium-labeled residues corresponding to the specific amino acids that interact with the antibody. See, e.g., Ehring (1999) Analytical Biochemistry 267: 252-; engen and Smith (2001) anal. chem.73: 256A-265A.
Modification assisted analysis (MAP), also known as antigen structure based antibody analysis (ASAP), is a method of classifying a large number of monoclonal antibodies (mabs) against the same antigen based on the similarity of each antibody's binding pattern to the surface of a chemically or enzymatically modified antigen (see US 2004/0101920, incorporated herein by reference in its entirety). Each class may reflect a unique epitope that is distinct from or partially overlapping with an epitope represented by another class. This technique allows rapid filtration of genetically identical antibodies, allowing characterization to focus on genetically different antibodies. When used in hybridoma screening, MAP may be helpful in identifying rare hybridoma clones that produce mabs with desired properties. MAP can be used to classify antibodies of the invention into groups of antibodies that bind different epitopes.
The present disclosure provides anti-Cx 43 antibodies that bind to the same epitope or a portion of an epitope. Likewise, the disclosure also includes anti-Cx 43 antibodies that compete for binding to Cx43 or fragment thereof with any of the specific exemplary antibodies described herein. For example, the disclosure includes anti-Cx 43 antibodies that cross-compete with one or more antibodies obtained from those described herein for binding to Cx 43.
Whether an antibody binds to the same epitope or competes for binding with a reference anti-Cx 43 antibody can be readily determined by using conventional methods known in the art. For example, to determine whether a test antibody binds to the same epitope as a reference anti-Cx 43 antibody of the invention, the reference antibody can be allowed to bind to Cx43 or a peptide under saturating conditions. Next, the ability of the test antibody to bind to the Cx43 molecule was evaluated. If the test antibody is capable of binding Cx43 after saturation binding of the reference anti-Cx 43 antibody, it can be concluded that the test antibody binds to a different epitope than the reference anti-Cx 43 antibody. On the other hand, if the test antibody is unable to bind to Cx43 after saturating binding with the reference anti-Cx 43 antibody, the test antibody can bind to the same epitope as the reference anti-Cx 43 antibody of the present disclosure.
To determine whether an antibody competes for binding with a reference anti-Cx 43 antibody, the above binding method can be performed in two directions: in a first orientation, a reference antibody can be allowed to bind to Cx43 under saturating conditions, and then the binding of the test antibody to the Cx43 molecule is assessed. In the second orientation, the test antibody can be allowed to bind to Cx43 under saturating conditions, and then the binding of the reference antibody to the Cx43 molecule is assessed. If only the first (saturating) antibody is able to bind to Cx43 molecule in both directions, then it can be concluded that the test and reference antibodies compete for binding to Cx 43. As will be understood by one of ordinary skill in the art, an antibody that competes for binding with a reference antibody may not necessarily bind the same epitope as the reference antibody, but may sterically block binding of the reference antibody by binding to an overlapping or adjacent epitope.
Two antibodies bind to the same or overlapping epitopes if each competitively inhibits (blocks) the binding of the other antibody to the antigen. That is, a1, 5, 10, 20 or 100 fold excess of one antibody inhibits the binding of the other antibody by at least 50%, but preferably 75%, 90% or even 99% as measured in a competitive binding assay (see, e.g., Junghans et al, Cancer Res.199050: 1495-. Alternatively, two antibodies have the same epitope if substantially all of the amino acid mutations in the antigen that reduce or eliminate binding of one antibody reduce or eliminate binding of the other antibody. Epitopes of two antibodies overlap if certain amino acid mutations that reduce or eliminate binding of one antibody reduce or eliminate binding of the other antibody.
Other routine experiments (e.g., peptide mutation and binding analysis) can then be performed to confirm whether the observed lack of binding of the test antibody is actually due to binding to the same epitope as the reference antibody or whether steric blockade (or other phenomena) is responsible for the lack of observed binding. Such experiments can be performed using ELISA, RIA, surface plasmon resonance, flow cytometry or any other quantitative or qualitative antibody binding assay available in the art.
In another aspect, provided herein are antibodies that partially or fully bind to an epitope located within amino acid sequence FLSRPTEKTI (SEQ ID NO: 19). In some embodiments, an epitope may comprise one or more amino acids selected from the group consisting of: f1, S3, R4, P5, T6, E7, K8, T9 and I10 of SEQ ID NO 19. In one embodiment, the epitope consists of F1, S3, R4, P5, T6, E7, K8, T9, and I10 of SEQ ID NO 19. In some embodiments, the epitope may include all 10 amino acids of SEQ ID NO 19. In some embodiments, an epitope may consist of all 10 amino acids of SEQ ID NO 19.
Pharmaceutical composition and use thereof
In another aspect, there is provided a pharmaceutical composition useful in the methods disclosed herein, i.e., for promoting the opening of Cx43 hemichannels in bone cells, preferably for the treatment of cancer, cancer metastasis, osteosarcoma, osteoporosis or osteopenia.
In some embodiments, the pharmaceutical composition comprises a Cx43 ligand and a pharmaceutically acceptable carrier. The Cx43 ligand can be formulated into pharmaceutical compositions with a pharmaceutically acceptable carrier. In addition, the pharmaceutical composition may include instructions for, e.g., using the composition to treat a patient to promote opening of Cx43 hemichannels in bone cells, preferably for treating cancer, cancer metastasis, osteosarcoma, osteoporosis, or osteopenia.
In one embodiment, the Cx43 ligand can be an anti-Cx 43 antibody or antigen-binding fragment thereof.
As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, and other excipients that are physiologically compatible. Preferably, the carrier is suitable for parenteral, oral or topical administration. Depending on the route of administration, the active compound (e.g., a small molecule or biologic) may be encapsulated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound.
Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions, and the conventional excipients used in the preparation of tablets, pills, capsules, and the like. The use of such media and agents in the formulation of pharmaceutically active substances is known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, it is contemplated that it will be used in the pharmaceutical compositions provided herein. Supplementary active compounds may also be incorporated into the compositions.
The pharmaceutically acceptable carrier may include a pharmaceutically acceptable antioxidant. Examples of pharmaceutically acceptable antioxidants include: (1) water-soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium hydrogensulfate, sodium metabisulfite, sodium sulfite, and the like; (2) oil-soluble antioxidants such as ascorbyl palmitate, Butylated Hydroxyanisole (BHA), Butylated Hydroxytoluene (BHT), lecithin, propyl gallate, α -tocopherol, and the like; and (3) metal chelating agents such as citric acid, ethylenediaminetetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
Examples of suitable aqueous and nonaqueous carriers that can be employed in the pharmaceutical compositions provided herein include water, ethanol, polyols (e.g., glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants, when required. In many cases, it is useful to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. The inclusion of agents that delay absorption such as monostearate salts and gelatin in the composition can prolong the absorption of the injectable composition.
These compositions may also contain functional excipients such as preservatives, wetting agents, emulsifying agents and dispersing agents.
The therapeutic compositions should generally be sterile, pyrogen free and stable under the conditions of manufacture and storage. The compositions may be formulated as solutions, microemulsions, liposomes or other ordered structures suitable for high antibody concentrations.
Sterile injectable solutions can be prepared by incorporating the active compound and one or a combination of the above ingredients in the required amount in an appropriate solvent with sterilization by, for example, filtration. Generally, the active activator is incorporated into a sterile vehicle containing a basic dispersion medium and the other desired ingredients described above to prepare a dispersion. In the case of sterile powders for the preparation of sterile injectable solutions, the methods of preparation include vacuum drying and freeze-drying (lyophilization) that yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The active agent may be mixed under sterile conditions with other pharmaceutically acceptable carriers, and with any preservatives, buffers, or propellants which may be required.
Prevention of the appearance of microorganisms can be ensured by a sterilization step (see above) and the addition of various antibacterial and antifungal agents, for example parabens, chlorobutanol, phenol, sorbic acid, and the like. Isotonic agents, such as sugars, sodium chloride, and the like may also be included in the compositions as desired. In addition, prolonged absorption of the injectable pharmaceutical form can be brought about by the addition of substances which delay absorption, for example, aluminum monostearate and gelatin.
Pharmaceutical compositions comprising Cx43 ligands can be administered alone or in combination therapy. For example, a combination therapy can include a composition provided herein that includes a Cx43 ligand and at least one or more other therapeutic agents, such as one or more chemotherapeutic agents known in the art, as will be discussed in further detail below. The pharmaceutical composition may also be administered in combination with radiation therapy and/or surgery.
The dosage regimen is adjusted to provide the optimum desired effect (e.g., therapeutic effect). For example, it may be a bolus injection, administered in time-divided doses or the dose may be reduced or increased in proportion to the urgency of the treatment.
Exemplary dosage ranges for administering the antibody include: 10-1000mg (antibody)/kg (patient weight), 10-800mg/kg,10-600mg/kg,10-400mg/kg,10-200mg/kg,30-1000mg/kg,30-800mg/kg,30-600mg/kg,30-400mg/kg,30-200mg/kg,50-1000mg/kg,50-800mg/kg,50-600mg/kg,50-400mg/kg,50-200mg/kg,100-1000mg/kg,100-900mg/kg,100-800mg/kg,100-700mg/kg,100-600mg/kg,100-500mg/kg,100-400mg/kg,100-300mg/kg and 100-200 mg/kg. Exemplary dosage schedules include once every three days, once every five days, once every seven days (i.e., once a week), once every ten days, once every fourteen days (i.e., once every two weeks), once every twenty-one days (i.e., once every three weeks), once every 28 days (i.e., once every four weeks), and once a month.
It is advantageous to formulate parenteral compositions in unit dose to facilitate administration and uniformity of dosage. A unit dose, as used herein, refers to physically discrete units administered as single doses to a patient to be treated, each unit containing a predetermined quantity of active agent calculated to produce any desired therapeutic effect, in association with a desired pharmaceutical carrier. The specification for the unit dosage form of the invention depends on or directly depends on (a) the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding the sensitivity of such active compounds for treatment in individuals.
Actual dosage levels of the active ingredients in the pharmaceutical compositions disclosed herein can be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition and mode of administration, and which is non-toxic to the patient. For administration, the term "parenteral administration" as used herein means forms of administration other than enteral and topical administration, typically by injection, including, but not limited to, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraocular, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular (subacosulalar), subarachnoid, intraspinal, epidural, and intrasternal injection and infusion.
The terms "parenteral administration" and "parenterally administered" as used herein refer to modes of administration other than enteral (i.e., through the alimentary canal) and topical administration, typically by injection or infusion, including, but not limited to, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subconjunctival, subarachnoid, intraspinal, epidural and intrasternal injection and infusion. Intravenous injection and infusion are commonly used (but not limited to) for antibody administration.
When the agents provided herein are administered as medicaments to humans or animals, they may be administered alone or as a pharmaceutical composition containing, for example, from 0.001 to 90% (e.g., from 0.005 to 70%, e.g., from 0.01 to 30%) of the active ingredient in combination with a pharmaceutically acceptable carrier.
In certain embodiments, the methods and uses provided herein for promoting the opening of Cx43 hemichannels in bone cells, preferably for treating cancer, cancer metastasis, osteosarcoma, osteoporosis, or osteopenia, can comprise administering a Cx43 ligand and at least one other anti-cancer agent that is not a Cx43 ligand.
In one embodiment, the at least one additional anti-cancer agent comprises at least one chemotherapeutic drug. Non-limiting examples of such chemotherapeutic drugs include platinum-based chemotherapeutic drugs (e.g., cisplatin, carboplatin), taxanes (e.g., paclitaxel)
Figure BDA0002716162770000271
Docetaxel
Figure BDA0002716162770000272
EndoTAG-1TM(formulation of paclitaxel encapsulated in a positively charged lipid complex; MediG)ene),
Figure BDA0002716162770000273
(paclitaxel preparation bound to albumin)), a tyrosine kinase inhibitor (e.g., imatinib @)
Figure BDA0002716162770000274
Sunitinib-
Figure BDA0002716162770000275
Dasatinib-
Figure BDA0002716162770000276
) And combinations thereof.
In another embodiment, the at least one additional anti-cancer agent comprises an EGFR inhibitor, e.g., an anti-EGFR antibody or a small molecule inhibitor of EGFR signaling. An exemplary anti-EGFR antibody is cetuximab
Figure BDA0002716162770000277
Cetuximab is available from Imclone Systems Incorporated. Other examples of anti-EGFR antibodies include matuzumab (EMD72000), panitumumab (R
Figure BDA0002716162770000278
Amgen); nimotuzumab (TheraCIM)TM) And mAb 806. An exemplary small molecule inhibitor of the EGFR signaling pathway is gefitinib
Figure BDA0002716162770000279
It is commercially available from astrazene and Teva. Other examples of small molecule inhibitors of the EGFR signaling pathway include erlotinib hydrochloride (OSI-774;
Figure BDA00027161627700002710
OSI Pharma); lapatinib (A)
Figure BDA00027161627700002711
GlaxoSmithKline); cantinib (cantinib dihydrochloride, Pfizer); pelitinib (Pfizer)(ii) a PKI-166 (Nowa); PD 158780; and AG 1478(4- (3-chlororonino) -6, 7-dimethoxyquinazoline).
In yet another embodiment, the at least one additional anti-cancer agent comprises a VEGF inhibitor. Exemplary VEGF inhibitors include anti-VEGF antibodies, e.g., bevacizumab (r) ((r))
Figure BDA00027161627700002712
Genentech)。
In yet another embodiment, the at least one additional anti-cancer agent comprises an anti-ErbB 2 inhibitor. Suitable anti-ErbB 2 antibodies include trastuzumab and pertuzumab.
In one aspect, the improved efficacy of a combination according to the present disclosure can be demonstrated by achieving therapeutic synergy.
The term "therapeutic synergy" is used when the combination of two products at a given dose is more optimal than each of the two products at the same dose. In one example, treatment synergy can be assessed by comparing the combination to the optimal single agent using estimates obtained from a two-way analysis of variance for repeated measurements of the parameter tumor volume (e.g., time factor).
The term "additive" means that the combination of two or more products at a given dosage is equivalent to the sum of the effects achieved by each of the two or more products, while the term "super-additive" means that the combination is more effective than the sum of the effects achieved by the two or more products.
Another way in which potency (including combined potency) can be quantified is by calculating log10A rate of cell killing, the rate of killing determined according to the following formula: log (log)10Cell killing ═ T-C (day)/3.32 xtdWherein T-C represents the delay in cell growth, which is the average time (days) for the treated (T) and control (C) tumors to reach a predetermined value (e.g., 1g or 10ml), and TdRepresents the time, in days, required for the tumor volume to double in control animals. When this measure is applied, if log10The product is considered to be active if the cytocidal power is greater than or equal to 0.7; if log10High cytocidal powerAt 2.8, the product is considered very active.
Using this measurement, the combination used at its own maximum tolerated dose (where each ingredient is generally present at a dose less than or equal to its maximum tolerated dose) is log10Cell killing greater than log when best component is administered alone10At the cell kill value, a therapeutic synergy is shown. Log of combinations in an exemplary case10Log of cell killing value over optimal component of combination10Cell killing value at least one log cell killing value.
Disclosed herein are compositions and methods for providing cancer therapy. The method can include promoting opening of a Cx43 hemichannel in bone cells in a subject in need thereof. Cx43 modulation (e.g., anti-Cx 43 antibodies) can be used as a stand-alone cancer treatment or in combination with other cancer treatments.
Also provided herein is a method for promoting the opening of a Cx43 hemichannel in an osteocyte, preferably for treating cancer, cancer metastasis, osteosarcoma, osteoporosis or osteopenia, comprising administering any one or more of the anti-Cx 43 antibodies disclosed herein in a subject in need thereof.
In various embodiments, the methods disclosed herein can comprise administering to the subject an effective amount of an anti-Cx 43 antibody or antigen-binding fragment thereof. Generally, an effective amount can be administered therapeutically and/or prophylactically.
The treatment may suitably be administered to a subject, particularly a human, suffering from, susceptible to or at risk of such cancer. Those subjects "at risk" can be arbitrarily objectively or subjectively determined by diagnostic tests or comments (e.g., genetic tests, enzyme or protein markers, family history, etc.) of the subject or health care provider. Identifying a subject in need of such treatment may depend on the judgment of the subject or a health care professional, and may be subjective (e.g., opinion) or objective (e.g., measurable by testing or diagnostic methods).
Administration of the formulation
The formulations of the present disclosure, including but not limited to reconstituted and liquid formulations, can be administered to a mammal, preferably a human, in need of treatment with an anti-Cx 43 antibody according to known methods, e.g., as a bolus intravenous bolus injection, or by continuous infusion over a period of time, or by intramuscular, intraperitoneal, intraspinal, subcutaneous, intraarticular, intrasynovial, intrathecal, oral, topical, or inhalation routes.
In embodiments, the formulation is administered to the mammal by intravenous or subcutaneous (i.e., under the skin) administration. For this purpose, the preparation can be injected using a syringe. However, other means of administering the formulation may be used, such as injection means (e.g. INJET-EASE TM and GENJET)TMA device); sample injection pen (e.g. GENPEN)TM) (ii) a Auto-injector devices, needleless devices (e.g. MEDIJECTOR)TMAnd BIOJECTORTM) (ii) a And a subcutaneous patch delivery system.
In a specific embodiment, the present disclosure relates to a kit for a single dose administration unit. Such kits comprise a container for an aqueous formulation of a therapeutic protein or antibody, including a single or multi-chamber prefilled syringe. An exemplary pre-filled syringe is available from Vetter GmbH of Ravensburg, germany.
The appropriate dosage of the protein ("therapeutically effective amount") will depend, for example, on the condition to be treated, the severity and course of the condition, whether the protein is administered for prophylactic or therapeutic purposes, previous treatment methods, the patient's clinical history and response to anti-Cx 43 antibody, the form of the formulation used, and the discretion of the attending physician. The anti-Cx 43 antibody can suitably be administered to a patient at once or over a series of treatments, and can be administered to the patient at any time from the start of diagnosis. anti-Cx 43 antibodies can be used alone or in combination with other drugs or therapies useful for treating the disease.
For anti-Cx 43 antibodies, the initial candidate dose for administration to a patient may be in the range of about 0.1-100 or 1-20mg/kg, which may take the form of one or more separate administrations. However, other dosage regimens may be useful. The progress of such treatment can be readily monitored by conventional techniques.
According to certain embodiments of the present disclosure, multiple doses of the anti-Cx 43 antibody (or a pharmaceutical composition comprising a combination of the anti-Cx 43 antibody and any other therapeutically active agent mentioned herein) may be administered to the subject within a determined period of time. Methods according to this aspect of the disclosure include sequentially administering multiple doses of the anti-Cx 43 antibodies of the disclosure to a subject. As used herein, "sequential administration" refers to administration of each dose of anti-Cx 43 antibody to a subject at different time points (e.g., on different days separated by predetermined intervals (e.g., hours, days, weeks, or months)). The present disclosure includes a method comprising: the patient is sequentially administered a single initial dose of anti-Cx 43 antibody, followed by one or more sequential doses of anti-Cx 43 antibody, and optionally followed by one or more subsequent doses of anti-Cx 43 antibody. The anti-Cx 43 antibody can be administered at a dose of between 0.1mg/kg to about 100 mg/kg.
The terms "initial dose", "second dose", and "third dose" refer to the temporal order of administration of the anti-Cx 43 antibodies of the invention. Thus, an "initial dose" is a dose administered at the beginning of a treatment regimen (also referred to as a "baseline dose"); "second dose" is the dose administered after the initial dose; the "third dose" is the dose administered after the second dose. Initially, both the second and third doses may contain the same amount of anti-Cx 43 antibody, but may typically differ from each other in dosing frequency. However, in certain embodiments, the amount of anti-Cx 43 antibody contained in the initial, second and/or third doses are different from each other (e.g., adjusted up or down as appropriate) during the course of treatment. In certain embodiments, two or more (e.g., 2, 3, 4, or 5) doses are administered at the start of a treatment regimen at a "loading dose" followed by administration of subsequent doses at a lower frequency (e.g., a "maintenance dose").
In certain exemplary embodiments of the present disclosure, each second and/or third dose is administered on weeks 1 through 26 (e.g., 1, 11/2, 2, 21/2, 3, 31/2, 4, 41/2, 5, 51/2, 6, 61/2, 7, 71/2, 8, 81/2, 9, 91/2, 10, 101/2, 11, 111/2, 12, 121/2, 13, 131/2, 14, 141/2, 15, 151/2, 16, 161/2, 17, 171/2, 18, 181/2, 19, 191/2, 20, 201/2, 21, 211/2, 22, 221/2, 23, 231/2, 24, 241/2, 25, 251/2, 26, 261/2, etc.) following administration of the immediately preceding dose. As used herein, the phrase "immediately preceding dose" refers to a dose of anti-Cx 43 antibody in a sequence of multiple administrations, administered to a patient without intermediate intervention prior to the administration of the next dose in the sequence.
Methods according to this aspect of the disclosure can include administering any number of the second and/or third doses of the anti-Cx 43 antibody to the patient. For example, in certain embodiments, only a single second dose is administered to the patient. In other embodiments, two or more (e.g., 2, 3, 4,5, 6,7, 8, or more) second doses are administered to the patient. For example, in certain embodiments, only a single third dose is administered to the patient. In other embodiments, two or more (e.g., 2, 3, 4,5, 6,7, 8, or more) third doses are administered to the patient.
In embodiments involving multiple second doses, each second dose may be administered at the same frequency as the other second doses. For example, each second dose may be administered to the patient 1-2 weeks or 1-2 months after the immediately preceding dose. Similarly, in embodiments involving multiple third doses, each third dose may be administered at the same frequency as the other third doses. For example, each third dose may be administered to the patient 2-12 weeks after the immediately preceding dose. In certain embodiments of the present disclosure, the frequency of administration of the second and/or third dose to the patient may vary over the course of the treatment regimen. The frequency of administration can also be adjusted during the course of treatment by the physician to the needs of the individual patient after clinical examination.
The present disclosure includes administration regimens in which a-10 or 2-6 loading dose is administered to a patient at a first frequency (e.g., once per week, once every two weeks, once every three weeks, once per month, once every two months), followed by two or more maintenance doses given to the patient at a lower frequency. For example, according to this aspect of the disclosure, if the loading dose is administered at a frequency, e.g., once a month (e.g., twice, three, four or more loading doses per month), the maintenance dose can be administered to the patient once every five weeks, once every six weeks, once every seven weeks, once every eight weeks, once every ten weeks, once every twelve weeks, etc.
Examples
The following examples, including experiments conducted and results obtained, are for illustrative purposes only and should not be construed as limiting the present disclosure.
Example 1: binding affinity
The optimized sequences were tested for binding affinity to Cx43 according to the following protocol.
1. Desorption Biacore T200
2. Inserting a new CM5 chip
Prime 3X and HBS-EP + buffers (10mM heparin, 150mM NaCl,3mM EDTA, 0.05% surfactant P20[ Tween20 ])
4. The preconditions are as follows: a new sensorgram was started at a rate of 100 ul/min. 10ul of 2X 100mM HCl, 2X 50mM NaOH, 2X 0.5% SDS in highly viscous solution (ultra-clean) were injected separately with REGEN COMMAND. This will clean and prepare the chip for coupling. This operation is performed only on blank chips, not after attachment of proteins.
5. Amine-coupled anti-human IgG Fc (GE: BR-1008-39) was used with each surface that you would use (including the reference surface) according to the instructions attached to the GE human antibody capture kit. Briefly, monoclonal antibodies were diluted to 25ug/ml in 10mM sodium acetate (pH 5). A new sensorgram was started at a rate of 5 ul/min. 7' was rapidly activated with NHS/EDC, Mab 7' was injected and blocked with ethanolamine 7 '. Typically 10,000-12,000 RU/surface are obtained. Then using 3M MgCl at a rate of 20ul/min210 regenerations were carried out for 30 seconds each. (Note that experiments with mouse IgG were similar, but we used the GE mouse antibody capture kit (GE: BR-1008-38.) the only difference was that the anti-mouse IgG antibody was coupled at a concentration of 30ug/ml and regenerated with 10mM glycine-HCl (pH 1.7) at 20# # # l/min for 3 minutes.
6. Normalization was then performed 1X Prime in HBS-EP + buffer.
7. The experiment was then performed immediately after amine coupling. At a significant time between experiments, the chips were removed and stored at 4 degrees celsius. After the chip was returned to the instrument, it was 3X primed (using HBS-EP +), calibrated, and primed with HBS-EP + 1X.
The program was written with the following parameters:
SUMMARY
Buffer solution HBS-EP +
Flow rate of 100ul/min
Data acquisition rate of 1Hz
The sample compartment temperature was 15 ℃ (this is the temperature the sample was maintained before injection).
The experimental operation temperature is 25 DEG C
Dual assay, Fc2-Fc1
For each experimental cycle:
a. mab Ab # K was captured on FC2 by injecting 5ug/ml Mab at 5ul/min for 180 seconds on FC2, with additional washing only after injection of 1% Tween 20.
b. 1M NaCl was injected over 30 seconds at 30ul/min on both FCs, followed by an additional buffer wash and stabilization for 180 seconds.
c. The sample (peptide) was injected by high-efficiency injection at 100ul/min for 210 seconds, dissociated on two FCs for 300 seconds, and then subjected to additional buffer washing and 60 seconds of stabilization.
e. With 3M MgCl2Both surfaces were regenerated at 20ul/min for 30 seconds, a high viscosity solution was selected, followed by buffer washing and 60 seconds of stabilization.
The loop programming is as follows:
1. buffer was injected 10 times initially to stabilize the instrument.
2. Concentration series for each peptide (PEP1, PEP2, PEP 3): 0,4nM,12nM,37nM,111nM,333nM,1000nM
Cycle 1-10 initial
Loop 11-17PEP1(914)
Loop 18-24PEP1(915)
Loop 25-31PEP1(916)
Cycle 32-38PEP1(914)
Loop 39-45PEP1(915)
Loop 46-52PEP1(916)
Loop 53-59PEP1(914)
Loop 60-66PEP1(915)
Loop 67-73PEP1(916)
Data analysis
Data were analyzed using T200 evaluation software 2.0. Since equilibrium is reached under optimal kinetic conditions, the data for each triplicate (Fc2-Fc1) generally fit the 1:1 binding kinetic model or steady state affinity model. The results obtained for both methods were similar.
For all experiments in which monoclonal antibodies were captured on the surface of a CM5 chip, the protocol described above was followed.
CAP chips were also used according to the following protocol:
1. CAP chips in the Biotin CAPture kit from GE Healthcare (28920234) were prepared according to the manufacturer's instructions. Briefly, it was connected in the instrument (T200), perfused three times with running buffer (HBS-EP +) and hydrated overnight with running buffer in standby mode. Then adjusted by injection with regeneration solution (6M GuHCl, 250mM NaOH) at 30ul/min for 3X 60 seconds. Then scaling 1 time, pre-run 1 time. It is then ready for the experiment. The program was written with the following parameters:
SUMMARY
Buffer solution HBS-EP +
Flow rate of 100ul/min
Data acquisition rate of 1Hz
The sample compartment temperature was 15 ℃ (this is the temperature the sample was maintained before injection).
The experimental operation temperature is 25 DEG C
Dual assay, Fc4-Fc3
For each experimental cycle:
a. biotin capture reagents were captured on Fc3 and Fc4 by injecting biotin capture reagents from the kit at a rate of 2 ul/min for 300 seconds.
b. Biotinylated peptide was captured on Fc4 by injecting 3ug/ml peptide 2120 sec at 5ul/min, followed by additional buffer washes and 120 sec stabilization.
c. Samples (monoclonal antibodies) were injected at 100ul/min for 210 seconds and dissociated for 300 seconds on both Fc.
d. Both surfaces were regenerated with 6M GuHCl, 250mM NaOH at 30ul/min for 120 seconds (high viscosity solutions were selected), followed by buffer washing and stabilization for 120 seconds.
The loop programming is as follows:
1. buffer was injected 5 times initially to stabilize the instrument.
2. Serial concentrations of each Mab (I or H) were run at 0,6.2nM,18.5nM,55.6nM,166.7nM,500 nM.
Cycle 1-5 Start
Circulating 6-11 monoclonal antibody I
Circulating 12-17 monoclonal antibody H
Circulating 18-23 monoclonal antibody I
Circulating 24-29 monoclonal antibody H
The binding affinity results (table 1) indicate that binding affinity is generally at least maintained and in many cases, surprisingly increased.
Table 1: binding affinities of various antibodies
Figure BDA0002716162770000341
Figure BDA0002716162770000351
Example 2 Fc receptor binding assay
Fc effector function is mediated by binding of Fc to a receptor. Receptors include FCRI, fcria, FCRIIb, FCRIIa, FCRIIb, Clq and FcRn. It is generally desirable to reduce binding affinity to most Fc receptors, with the expectation that potential in vivo toxicity of FcRn is minimized while maintaining antibody half-life. The following Surface Plasmon Resonance (SPR) and enzyme-linked immunosorbent assay (ELISA) protocols were used to test the binding of various antibodies to different Fc receptors.
Fcri binding
Experiment: biacore 8K
Chip: CM5
(1) Immobilization of
The activator was prepared by mixing 400mM EDC and 100mM NHS immediately before injection. The mixture activates the CM5 sensor chip for 420 seconds. Then 30. mu.g/mL of THE in 10mM NaAc (pH 4.5)TMHis-tag antibody was injected into channels 1-8 at 30. mu.L/min for 400 seconds. The chip was deactivated with 1M ethanolamine-HCl (GE).
(2) Capturing ligands and running analytes
2 μ g/mL CD64 in running buffer (1xHBS-EP +) was injected at 10 μ L/min into Fc2 for 30 seconds in channels 1-4. 6 concentrations (40, 20, 10, 5, 2.5 and 1.25nM) of analytes 20170905-Ab # C-02, 20170905-Ab # D-02, 20170908-Ab # G-02, 20170920-Ab # H-02 and running buffer were injected sequentially at 30 μ L/min into Fc1 to Fc2 of channels 1-4, with a binding phase of 180, and then dissociated to 400. The cycle of 6 capture ligands and running analyte was repeated in ascending order according to analyte concentration. 10mM glycine, pH 1.5, was injected as regeneration buffer after each dissociation phase.
2 μ g/mL CD64 in running buffer (1xHBS-EP +) was injected at 10 μ L/min into Fc2 for 30 seconds in channels 1-6. The 8 concentrations (10240,5120,2560,1280,640,320,160 and 80nM) of analyte 20170907-Ab # K-02,20170908-Ab # L-02,20170915-Ab # O-02,20170919-Ab # P-02,20170919-Ab # S-02 and 20170920-Ab # T-02 and running buffer were injected sequentially at 30 μ L/min into Fc1 to Fc2 of channels 1-6, with a binding phase of 60, followed by dissociation to 90. The cycle of 8 capture ligands and running analyte was repeated in ascending order according to analyte concentration. 10mM glycine, pH 1.5, was injected as regeneration buffer after each dissociation phase.
(3) Regeneration
The chip was regenerated with 10mM glycine pH 1.5.
(4) Data analysis
The surface channel Fc1 without capture ligand was used as a control surface for reference subtraction (reference subtraction). The final data for each interaction is subtracted from the reference channel and buffer channel data. By 1:1 binding Pattern Experimental data for binding of 20170905-Ab # C-02, 20170905-Ab # D-02, 20170908-Ab # G-02, 20170920-Ab # H-02 to CD64 were fitted. The 10240nM curves for analytes 20170907-Ab # K-02,20170908-Ab # L-02,20170915-Ab # O-02,20170919-Ab # P-02,20170919-Ab # S-02 and 20170920-Ab # T-02 were removed for a better fit. The relative experimental data were fitted with steady-state affinity and are shown in table 2 below.
Table 2: FCRI bonding
Figure BDA0002716162770000361
All antibodies showed low or no FCRI binding, which is advantageous.
A. Binding to Fc γ RIIa, Fc γ RIIb, Fc γ RIIIa, Fc γ RIIIb
Experiment: biacore 8K
Chip: CM5
(1) Immobilization of
The activator was prepared by mixing 400mM EDC and 100mM NHS immediately before injection. The mixture activates the CM5 sensor chip for 420 seconds. Then 30. mu.g/mL of THE in 10mM NaAc (pH 4.5)TMHis-tag antibody was injected into channels 1-8 at 30. mu.L/min for 400 seconds. The chip was deactivated with 1M ethanolamine-HCl (GE).
(2) Capturing ligands and running analytes
mu.g/mLFc. gamma. RIIa, Fc. gamma. RIIb, Fc. gamma. RIIIa or Fc. gamma. RIIIb in running buffer (1XHBS-EP +) were injected into Fc2 of channels 1-8 at a flow rate of 10. mu.L/min for 15 seconds. Analytes are injected into channels 1-8, respectively. A series of analyte concentrations were monitored at a flow rate of 30 μ L/min during the association phase of 60 seconds followed by dissociation of 90 seconds (see table 3 below). 10mM glycine, pH 1.5, was injected as regeneration buffer after each dissociation phase.
Table 3: concentration of analyte
Analyte Concentration tested (nM)10
Ab#D 0,160,320,640,1280,2560,5120,10240
Others 0,320,640,1280,2560,5120,10240,20480,40960
(3) Regeneration
The chip was regenerated with 10mM glycine pH 1.5.
(4) Data analysis
The surface channel Fc1 without capture ligand was used as a control surface for reference subtraction (reference subtraction). The final data for each interaction is subtracted from the reference channel and buffer channel data. Experimental data for antibodies binding to Fc γ RIIa, Fc γ RIIb, Fc γ RIIIa and Fc γ RIIIb were fitted by steady state affinity patterns and are shown in table 4 below.
Table 4: fc γ RIIa, Fc γ RIIb, Fc γ RIIIa, and Fc γ RIIIb binding
Figure BDA0002716162770000371
Figure BDA0002716162770000381
All antibodies showed low or no Fc γ RIIa, Fc γ RIIb, Fc γ RIIIa, and Fc γ RIIIb binding, which is advantageous.
C. Binding to FcRn
Experiment: biacore 8K
Chip: CM5
(1) Buffer exchange
Buffer exchange of human FcRn with running buffer (50mM Na) using desalting column according to instruction manual2HPO4、50mM NaH2PO4150mM NaCl, 0.05% Tween20, pH 6.0). The concentration was determined using Nanodrop.
(2) Immobilization of
Activators were prepared by mixing 400mM EDC and 100mM NHS (GE) immediately prior to injection. The CM5 sensor chip was activated with the mixture at a flow rate of 10 μ L/min for 420 seconds. Then 5. mu.g/mL of antibody in 10mM NaAc (pH5.5) was injected into the Fc2 of channels 1-8 at a flow rate of 10. mu.L/min for 60 seconds, respectively. The opposite Fc1 was blocked. The chip was inactivated with 1M ethanolamine-HCl (GE) at a flow rate of 10. mu.L/min for 420 seconds.
(2) Running analytes
The analyte FcRn is injected into channels 1-8, respectively. FcRn (0,93.75,187.5,375,750,1500,3000 and 6000nM) was monitored at 8 concentrations at a flow rate of 30 μ L/min during the association phase of 60 seconds followed by dissociation of 90 seconds. After each round of interaction analysis cycles, the sensor chip surface was regenerated with 1XPBS (pH7.4) at a flow rate of 10. mu.L/min for 30 seconds.
(3) Regeneration
The chip was regenerated with 1xPBS (pH 7.4).
(4) Data analysis
The surface channel Fc1 without immobilized antibody was used as a control surface for reference subtraction (reference subtraction). The final data for each interaction is subtracted from the reference channel and buffer channel data. The relative experimental data were fitted with a steady state affinity model and are shown in table 5 below.
Table 5: FcRn binding
Ligands KD(M)
Ab#D 2.25E-06
Ab#H 2.61E-06
Ab#L 2.60E-06
Ab#P 2.55E-06
Ab#T 2.56E-06
All antibodies showed ideally similar FcRn binding.
D. Binding of C1q by ELISA
The plate (Nunc) was coated with 3. mu.g/mL of antibody overnight at 4 ℃. After blocking and washing, C1q was subjected to a semilog titration in blocking buffer (600, 189.75, 60.01, 18.98, 6.00, 1.90, 0.60, 0.19, 0.06, and 0.02 μ g/mL) and incubated at room temperature for 2 hours. The plates were then washed and subsequently incubated with the secondary antibody sheep anti-human C1q Ab-HRP for 1 hour. After washing, TMB substrate was added and the interaction was stopped with 2M HCl. The absorbance at 450nm was read using a microplate reader (Molecular Device) and is shown in table 6 below.
Table 6: c1q binding
Antibodies KD
Ab#D 75nM
Ab#H Without bonding
Ab#L Weak bond
Ab#P Without bonding
Ab#T Without bonding
All antibodies showed low or no binding of C1q, which is advantageous.
Example 3 epitope mapping
To reconstitute the target molecular epitope, a peptide-based epitope mimic library was synthesized using solid phase Fmoc synthesis. Amino-functionalized polypropylene supports were obtained by grafting proprietary hydrophilic polymer formulations, then reacting N-hydroxybenzotriazole (HOBt) with t-butoxycarbonyl-hexamethylenediamine (BocHMDA) using Dicyclohexylcarbodiimide (DCC), followed by cleavage of the Boc-group using trifluoroacetic acid (TFA). Peptides were synthesized on amino-functionalized solid supports using standard Fmoc-peptide synthesis methods by a custom modified JANUS liquid handling station (Perkin Elmer).
The synthesis of the structural mimetics was accomplished using the chemical linker on scaffold (CLIPS) technique. CLIPS technology allows the structuring of peptides into single, bi-, tri-, sheet, helical folds and combinations thereof. The CLIPS template is coupled to a cysteine residue. The side chains of multiple cysteines in the peptide are coupled to one or two CLIPS templates. For example, a 0.5mM solution of P2 CLIPS (2, 6-bis (bromomethyl) pyridine) is dissolved in ammonium bicarbonate (20mM, pH 7.8)/acetonitrile (1: 3 (v/v)). This solution was added to the peptide array. CLIPS template will bind to the side chains of two cysteines present in the peptide in a peptide array (455 well plate, with 3 μ l wells) solid phase. Peptide(s)The array was gently shaken in solution for 30 to 60 minutes while it was completely covered in solution. Finally, the peptide arrays were used in excess of H2O-washed well and sonicated in PBS (pH 7.2) at 70 ℃ for 30 min in disruption buffer containing 1% SDS/0.1% 2,2' - (ethylenedioxy) diethylmercaptan and then in water for 45 min. Peptides carrying T3 CLIPS were prepared in a similar manner, but now using three cysteines.
Different sets of peptides were synthesized according to the following design. Note that the actual order of the peptides on some microcards is random.
Figure BDA0002716162770000401
The binding of the antibody to each synthetic peptide was tested in an ELISA. The peptide arrays were incubated with an anti-solution (overnight at 4 ℃). After washing, the peptide arrays were incubated with 1/1000 diluted appropriate antibody peroxidase conjugate (SBA; goat anti-human HRP conjugate, Southern Biotech) for 1 hour at 25 ℃. After washing, the peroxidase substrate 2,2' -azido-bis-3-ethylbenzothiazoline sulfonate (ABTS) and 20. mu.l/ml of 3% H were added2O2. After one hour, color development was measured. The color development was quantified with a Charge Coupled Device (CCD) -camera and image processing system.
The values obtained from the CCD camera ranged from 0 to 3000mAU, similar to a standard 96-well plate ELISA reader. The results are quantified and stored in a laboratory database. Occasionally, one well will contain a bubble, resulting in a false positive value, requiring manual inspection of the card, and recording any value caused by the bubble as 0.
To verify the quality of the synthetic peptides, a set of independent positive and negative control peptides were synthesized in parallel. Screening was performed with the commercially available antibodies 3C9 and 57.9 (see Posthumus et al (1990) J. Virol. 64: 3304-3309).
Figure 1 gives a graphical overview of the complete data set. The box plot here depicts each data set and indicates the mean ELISA signal, distribution and outliers in each data set. Depending on the experimental conditions (amount of antibody, blocking strength, etc.), different ELISA data distributions can be obtained. Specifically, the bottom and top of the box are the 25 th and 75 th percentiles of the data. The bar near the middle of the box is the 50 th percentile (median). Whiskers were in the 1.5 range between quantiles, indicating statistical abnormalities in the dataset (Mcgill et al, (1978) statisticians in the United states, 32: 12-16).
Antibodies were tested at high concentrations under high stringency conditions. The results are reported in fig. 2 and 3. Data were analyzed for each of the two peptide groups.
Analysis of the data recorded with the substituted variants of leader sequence FLSRPTEKTI indicated that many substitutions of any one residue in the sequence affected negatively, albeit to a different extent, and affected antibody binding (FIG. 2). Only one exception was seen-residue L2, which was intolerant to L2P and L2Y substitutions, but was insensitive to all other substitutions.
Analysis of the data recorded for the truncated variant of leader sequence FLSRPTEKTI indicated that the N-terminus of the sequence was favoured by the antibody (FIG. 3). Many constructs from the central part of FLSRPTEKTI are also well known.
In summary, antibody-single residue mutants and truncated variants were tested on peptide arrays comprising two types of peptide variants derived from leader sequence FLSRPTEKTI. The antibody produces detectable binding under high stringency conditions. Many substitutions in FLSRPTEKTI were not suitable for the antibody. Antibodies are stronger binding truncated constructs from the N-terminal part of the sequence.
Example 4 antibody stability
Antibody stability is an important factor affecting development, efficacy, production cost, and the like. After sequence optimization, key stability parameters were evaluated. Species profiles of various antibodies under acidic and high temperature conditions were tested. All antibodies showed improved stability.
SE-UPLC (size exclusion ultra high performance liquid chromatograph)
Preparation: PBS, pH 6.5 or 7.2
Concentration (mg/mL): 5.28,5.13,5.00,5.17,5.01,5.26,4.92,5.04,4.99,5.12 (all at about 5mg/mL)
Conditions are as follows: at room temperature, acid treatment, and storage at 4 deg.C for 1 week or at 40 deg.C for 1 week
mu.L of the sample was injected over 10 minutes into an ACQUITY UPLC Protein BEH SEC 200, 1.7 μm, 4.6X 150mm column at a flow rate of 0.3 mL/min. A mobile phase of 50mM sodium phosphate, 500mM NaCl, pH 6.2 was used. All antibodies showed the required stability under various pH, heat and storage conditions.
BrCE-SDS (reduced capillary electrophoresis-sodium dodecyl sulfate)
Preparation: PBS, pH 6.5,7.2,6.2
Concentration (mg/mL): 0.5
Conditions are as follows: at room temperature, acid treatment, and storage at 4 deg.C for 1 week or at 40 deg.C for 1 week
Samples were prepared in reducing labeling buffer and then submitted to the LabChip gxi system (PerkinElmer). All antibodies showed the required stability under various pH, heat and storage conditions.
Example 5 semi-channel opening experiment
In vitro experiments
The effect of the antibodies disclosed herein on the opening of a half channel can be tested in vitro using a dye uptake assay. The dye may be a fluorescent tracer dye (e.g., ethidium bromide or fluorescein).
In one example, a Fluid Flow Loop Apparatus (FFLA) (parallel plate flow chamber) or a variation thereof may be used. The FFLA mimics the dynamic fluid microenvironment in the bone to create Fluid Flow Shear Stress (FFSS). Cells were cultured in parallel plate flow chambers and exposed to a steady laminar flow.
The bone cells sense the mechanical stress that the FFSS creates in the bone cell lumen/tubule network. It has been proposed that bone fluid flow is driven by extravascular pressure and the cyclic mechanical load exerted by bone cells, and that the maximum physiological load is 8 to 30dyn/cm2. In certain aspects, the FFSS levels are within the physiological values reported in studies measuring fluid flow in bone in previous studies. The magnitude of the fluid shear stress can be varied by adjusting the column height of the flow circuit.
Experiments for assessing the function of a half-channel may use fluorescent tracer molecules that are small enough to pass through the half-channel pore. If the hemichannel is closed, the molecule will not pass through. If the hemichannel is open, the dye can pass through and cause the cell to fluoresce, allowing fluorescence to be quantified. When ethidium bromide binds to DNA, it fluoresces. Once the fluorescein is inside the cell, it fluoresces.
The dye transfer method may comprise exposing the cells to an extracellular fluorescent permeability tracer. Extracellular permeability tracers are molecules that remain outside the cell unless the cell membrane permeability is increased under certain conditions. In certain aspects, the mass of the tracer is less than 1, 2 or 3 kDa. In other aspects, the tracer will have a net charge. Such permeability tracers include, but are not limited to, the anionic dye fluorescein (LY; net charge ═ 1) and the cationic probe ethidium bromide (Etd; net charge ═ 1), iodopropylidine (PI; net charge ═ 2). Upon binding to DNA, EtBr fluorescence increases, thereby increasing contrast and making it more readily identifiable. In certain aspects, extracellular dye is removed at different time periods or after applying a stimulus to open the hemichannel, and the retained fluorescence intensity in each cell is quantified. In certain aspects, fluorescence intensity is quantified in the snapshot images.
Substances used to test for hemichannel opening in vitro assays include:
a cell or cell line expressing a hemichannel. Cells or cell lines expressing various connexin hemichannels can be obtained, isolated or engineered using methods and/or expression vectors known in the art.
Bone cells: primary bone cells isolated from animals (including mice, rats, rabbits, chickens) or the like or bone cell lines including, but not limited to, MLO-Y4 cells and the like.
Cancer cell: breast cancer cell lines: including ER, PR, HER and TP53 positive/negative cells (e.g., MD-MBA-231, MCF7, T47D or ZR 751). MDA-MB-231 is ductal carcinoma of the breast. The Py8119 breast tumor cell line was established from spontaneous breast tumors generated in C57Bl/6MMTV-PyMT female mice (a polyoma mid-T transgene driven by the mouse mammary tumor virus promoter). Expression of oncogenes (mid-polyoma T transgene) is driven by the mouse mammary tumor virus promoter.
Prostate cancer cell lines: including androgen receptor and 5 alpha-reductase positive/negative and androgen sensitive/insensitive cell lines (e.g., LNCaP-Rf, BM18, pRNA-1-1/ras, RC58T/hTERT, PPC-1, etc.).
Osteoblasts: MLO-a5 osteoblasts were used as controls because they expressed connexin 43, but they did not appear to open under alendronate stimulation.
Tracer molecules include, but are not limited to, fluorescein, ethidium bromide, evans blue, Alexa350, Alexa488, and Alexa 594.
Cx43(E2) Cx43(E2) antibodies are specific for Cx43 hemichannels. Cx43E2 binds to the second extracellular loop of the Cx43 hemichannel and prevents the hemichannel from opening.
The method of determining whether an antibody opens a hemichannel comprises one or more of the following steps:
(a) isolating, obtaining or generating a cell or cell line expressing a connexin. For example, primary bone cells can be isolated from bone ash. Other cell types can be isolated using other methods known in the art. In certain aspects, the skull bone cells are isolated from an animal (e.g., a 16-day embryonic chicken skull or a neonatal mouse). The animals were decapitated, the cranium cut and quickly immersed in 70% alcohol. The cranium was then placed in α MEM and washed several times with PBS. The cleaned bone was placed in fresh α MEM. The bone was cut into pieces and cut into 1.5 mm area sizes. The bone pieces may be treated with collagenase to remove soft tissue and osteoid, and then decalcified using EDTA. Finally, the bone cells are released from the bone mass by treatment with collagenase and vigorous stirring.
(b) Primary bone cells are isolated from long bones. Long bone osteocytes can be isolated from mice or rats of 2-3 weeks of age. For example, mice are given an excess of anesthetic and then are dislocated in their neck, decapitated and immersed in 70% ethanol. Separating the femur and tibia with the joint ends still intact. The legs were quickly immersed in 70% alcohol and then placed in α MEM. The legs in α MEM were washed with PBS. Most of the muscle was removed and separated from the tendon/ligament. The cleaned bone was placed in fresh α MEM. After all bones were cleaned, both ends of each bone were cut with a scalpel before flushing the bone marrow with PBS. The bone is cut into lengths of 1.5 to 2mm and treated with collagenase. In one example, the bone pieces were treated sequentially 9 times with collagenase to remove all other tissues and osteoids, and then decalcified using EDTA.
(c) Culturing the cell or cell line. For example, primary and/or osteolytic cell lines are cultured on collagen-coated plates and immersed in recording medium containing a permeability tracer (HCO 3- α -MEM medium without HECO3, buffered with HEPES).
(d) A test antibody is administered. The time required to contact the cultured cells with the test antibody.
(e) The absorption of the permeability tracer is determined. Uptake of the permeability tracer is determined by measuring the amount of tracer in the cell. In some aspects, time-lapse recording is used. The fluorescence of the regions of interest of the different cells can be recorded in a microscope equipped with an eclipse filter based on the fluorescence wavelength of the tracer or other probe used. In certain aspects, a fast-cooled digital camera captures images every 2 minutes and performs image processing using ImageJ software. The data collected can be plotted as the fold difference of the initial fluorescence and the fluorescence at the time of interest relative to the base fluorescence.
For snapshot images, cells can be exposed to the permeability tracer for 5-10 minutes, washed multiple times with PBS, and fixed with formaldehyde. In certain aspects, at least three photomicrographs of the fluorescent field are taken with a microscope. Image analysis was done by ImageJ software. The mean pixel density of random cells was measured.
The opening of connexin hemichannels can be confirmed, for example, by incubating bone cells with a Cx43(E2) antibody, a polyclonal antibody that specifically inhibits the Cx43 hemichannel, and the test antibody. If the test antibody opens a Cx43 hemichannel, the opening of this channel will be blocked by the Cx43(E2) antibody. To control the opening of Cx43 hemichannels, osteocytes are subjected to fluid shear stress and/or AD treatment, both of which are known to open hemichannels in osteocytes.
In a specific example, MLO-Y4 bone cells were treated with 20 μ M AD or test antibody in the absence or presence of 1 μ g/ml Cx43(E2) antibody. Ethidium bromide dye uptake was performed and quantified compared to untreated basal uptake levels. The assay is performed in the presence of calcium. Low calcium conditions can be used as controls (open hemichannels). Cx43(E2) antibodies can block the opening of osteocyte hemichannels induced by AD or test antibodies.
In vivo assay
In certain aspects, modulation of Cx43 in bone cells is determined by injecting a candidate agent into long bone and detecting the opening of hemichannels in bone cells using a fluorescent tracer dye (e.g., calcein or evans blue).
An example of an in vivo assay for analysis of hemichannels in bone cells uses mice or rats 3-4 months old. Animals were weighed. Test antibodies were introduced into the animals by Intraperitoneal (IP) injection. After 2-4 hours, the fluorescent tracer dye (i.e., evans blue, Alexa 594) was injected into the lateral tail vein of the animal or injected by IP. Note that: up to 1% of the animal's body weight (by volume) can be injected. In certain aspects, the animal is heated to dilate the tail vein prior to injection of the tail vein. After 2-4 hours, animals were sacrificed and the tibia and femur without muscle tissue were excised and washed several times with PBS. Bones were fixed in paraformaldehyde and decalcified in 14% EDTA solution at 4 ℃ for two weeks or at room temperature with constant stirring for 3-5 days. Bones were washed in PBS and soaked overnight in 30% sucrose in PBS before being embedded in OCT compound. The position of the bones is usually adjusted in the mold as desired. Frozen sections of 5 μm thickness were cut using a cryostat, rinsed with PBS and then fixed with 50% glycerol in PBS. Bone sections can be examined under a fluorescent microscope and Image J used to quantify the extent of bone cells in bone that have taken up the tracer dye.
The opening of the Cx43 hemichannel in osteocytes can be confirmed by mechanical loading on the tibia that opens the Cx43 hemichannel in osteocytes. This can serve as a positive control for the opening of hemichannels in bone cells in vivo. For negative controls, mice lacking Cx43 in bone cells were used. The mice were generated by crossing 10-kb DMP-1Cre and Cx43 flox mice.
Example 6 determination of cancer cell migration, viability and metastasis
In vitro experiments
Cancer cell migration experiments Cx43 hemichannels in bone cells were opened by administration of AD or FFSS. The open hemichannel allows various factors to be released into the medium, resulting in Conditioned Medium (CM). Factors released in AD or FFSS treated CM were determined to reduce cancer cell migration by soft agar and wound healing assays. Cancer cells treated with control CM showed normal migration. In contrast to anchorage-dependent growth, the soft agar assay is an anchorage-independent growth assay. Only cancer cells can grow on soft agar, and their growth on this substrate indicates the extent of proliferation of the cancer cells.
In certain embodiments, cancer cells (e.g., breast or prostate cancer) cells are incubated with CM and the proliferation, migration, and invasion of the cancer cells is determined.
Growth and viability of cancer cells can be determined using WST-1 (water soluble tetrazolium salt), viable cell count using trypan blue method, BrdU DNA incorporation and cell proliferation assays. For the WST-1 assay, cell proliferation was measured using a Synergy HT multimodal microtiter plate reader (Biotek) at an emission wavelength of 450 nm.
Cell migration assays are typically performed in a transmembrane membrane filter insert in 24-well tissue culture plates (BD Biosciences). The cross-pore membrane filter insert may be 6.5mm in diameter, 8 μm in pore size, and 10nm thick in polycarbonate membrane.
The invasion assay was performed in a BD Biocoat growth factor reduced Matrigel invasion chamber (BD Biosciences). Cancer cell lines were collected and resuspended in CM from bone cells with or without test antibody. A suspension of cancer cells is added to the upper side of the insert. Cells were incubated at 37 ℃ for various periods of time. Cells that did not migrate through the filter were removed, cells that migrated through the insert were fixed and stained with a Hema 3Stat Pack (Fisher Scientific). The number of cells migrating in 5 fields per insert was counted under a light microscope.
Breast cancer cell migration can be reduced when incubated in CM from bone cells treated with AD or FFSS or antibodies disclosed herein to stimulate Cx43 hemichannel opening. This inhibition of cancer cell migration is reduced when the bone cell Cx43 hemichannel is blocked by the E2 antibody. This reduction in cancer cell migration was not seen when incubated with CM collected in osteoblasts or treated directly with AD. The antibodies disclosed herein protect the opening of Cx43 hemichannels against the growth and migration of breast cancer cells.
A. In vivo assay
The effect of the test antibody on bone metastasis in vivo was determined using an intra-tibial injection bone metastasis model and/or an intra-cardiac injection cancer metastasis assay.
Tibia internal bone metastasis model: the method comprises anesthetizing a1 month old normal or immunocompromised mouse with isoflurane. Buprenorphine hydrochloride (0.3mg/ml) was also administered to mice as an analgesic. Cancer cells expressing fluorescent or chemiluminescent markers (e.g., Py8119 cells expressing Luc-GFP in normal mice or MD-MBA-231 expressing Luc-GFP in immunocompromised mice are injected intrapshinally. cancer cells are seeded into the bone marrow region of the right tibia by pre-perforation with a Hamilton syringe fitted with a 30 gauge needle. PBS is injected into the left tibia as a control. test antibody or saline is IP administered twice weekly for 5 weeks starting 3 days after tumor cell seeding, tumor growth in the tibia is monitored weekly by bioluminescence imaging or fluorescence.
Intracardiac bone metastasis model: two three month old normal or immunocompromised mice were anesthetized with isoflurane and buprenorphine hydrochloride (0.3mg/ml) was given as an analgesic. Cancer cells expressing a fluorescent or chemiluminescent marker (e.g., Py8119 cells expressing Luc-GFP in normal mice or Py8119 cells expressing Luc-GFP-MD-MBA-231 in immunocompromised mice) are injected into the left ventricle of the mice. The process comprises the following steps: the needle is held facing the operator and tilted to the right and inserted into the second intercostal space approximately 3mm to the left of the sternum. Approximately 5mm of advancement, then the needle was gently rotated until a pulsatile flow of bright red arterial blood was observed into the needle hub. The cell suspension was injected over 30 seconds. The needle was pulled out and the injection site was then pressed with an alcohol swab for 30 seconds. Mice were placed on a warm surface until recovery from anesthesia was complete. Following intracardiac injection, bioluminescence or fluorescence imaging was performed weekly from 3 days post tumor cell inoculation to verify tumor cell distribution. At the end of the study after sufficient bioluminescence imaging, X-ray images were taken to assess bone quality and labeled metastatic cancer cell colonies were observed and counted using a fluorescence microscope.
Cx43 conditional knockout (cKO) mice. Since the homozygous Cx43 global knockout was lethal, and since the inventors wished to examine the effects of Cx43 expressed in bone cells, bone cell specific Cx43 knockout mice were generated. Mice homozygous for floxed Cx43 gene were crossed with mice heterozygous for Cx43 as a whole to promote complete deletion of Cx43 in bone cells. Cx43f 1/-mice (50% of progeny) were then crossed with mice expressing Cre recombinase driven by the human DMP-1 promoter. The mice so produced were Cx43 fl/-, DMP 1Cre + or Cx43 fl/-, DMP1 Cre- (small ratio Cx43fl/fl or Cx 43-/-). Cx43 deficient bone cells were confirmed by immunohistochemistry.
The study may include 8 groups of mice: alendronate (AD) treated WT, AD-free WT, cKO treated with AD, AD-free cKO, Test Antibody (TA) treated WT, TA-free WT, TA-treated cKO and TA-free cKO. Mice were administered either AD or TA at 150. mu.g/kg body weight. Bone metastasis in KO can be expected to increase compared to WT mice treated with AD or TA. Bone metastasis should be similar between WT and knockout mice in the absence of AD or TA treatment.
Various aspects of the present disclosure may be used alone, in combination, or in a variety of configurations not specifically discussed in the embodiments described in the foregoing and is therefore not limited in its application to the details and arrangement of components set forth in the foregoing description or illustrated in the drawings. For example, aspects described in one embodiment may be combined in any manner with aspects described in other embodiments.
While specific embodiments of the subject disclosure have been discussed, the above description is illustrative and not restrictive. Many variations of the disclosure will become apparent to those skilled in the art upon reading the present specification. The full scope of the disclosure should be determined by reference to the claims and their full scope of equivalents, and to such variations.
Is incorporated by reference
All publications, patents, and patent applications cited in this specification are herein incorporated by reference in their entirety for all purposes to the same extent as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference.
Sequence listing
<110> Alamab Therapeutics GmbH (AlaMab Therapeutics, Inc.)
<120> connexin 43 antibody and uses thereof
<130> 172628-020301/PCT
<150> US 62/651,668
<151> 2018-04-02
<160> 39
<170> PatentIn version 3.5
<210> 1
<211> 8
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 1
Gly Tyr Thr Phe Thr Ser Tyr Tyr
1 5
<210> 2
<211> 8
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 2
Ile Asn Pro Ser Asn Ala Gly Thr
1 5
<210> 3
<211> 12
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 3
Thr Arg Glu Gly Asn Pro Tyr Tyr Thr Met Asn Tyr
1 5 10
<210> 4
<211> 11
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 4
Gln Ser Leu Leu Glu Ser Asp Gly Lys Thr Tyr
1 5 10
<210> 5
<211> 3
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 5
Leu Val Ser
1
<210> 6
<211> 9
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 6
Trp Gln Gly Thr His Phe Pro Trp Thr
1 5
<210> 7
<211> 119
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 7
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Tyr Met Tyr Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Gly Ile Asn Pro Ser Asn Ala Gly Thr Asn Phe Asn Glu Lys Phe
50 55 60
Lys Asn Arg Ala Thr Leu Thr Val Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Thr Arg Glu Gly Asn Pro Tyr Tyr Thr Met Asn Tyr Trp Gly Gln Gly
100 105 110
Thr Leu Val Thr Val Ser Ser
115
<210> 8
<211> 112
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 8
Asp Val Val Met Thr Gln Ser Pro Leu Ser Leu Pro Val Thr Ile Gly
1 5 10 15
Gln Pro Ala Ser Ile Ser Cys Lys Ser Ser Gln Ser Leu Leu Glu Ser
20 25 30
Asp Gly Lys Thr Tyr Leu Asn Trp Leu Gln Gln Arg Pro Gly Gln Ser
35 40 45
Pro Arg Arg Leu Ile Tyr Leu Val Ser Lys Leu Asp Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Trp Gln Gly
85 90 95
Thr His Phe Pro Trp Thr Phe Gly Gly Gly Thr Lys Val Glu Ile Lys
100 105 110
<210> 9
<211> 449
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 9
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Tyr Met Tyr Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Gly Ile Asn Pro Ser Asn Gly Gly Thr Asn Phe Asn Glu Lys Phe
50 55 60
Lys Asn Arg Ala Thr Leu Thr Val Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Thr Arg Glu Gly Asn Pro Tyr Tyr Thr Met Asn Tyr Trp Gly Gln Gly
100 105 110
Thr Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe
115 120 125
Pro Leu Ala Pro Ser Ser Lys Ser Thr Ser Gly Gly Thr Ala Ala Leu
130 135 140
Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp
145 150 155 160
Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu
165 170 175
Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser
180 185 190
Ser Ser Leu Gly Thr Gln Thr Tyr Ile Cys Asn Val Asn His Lys Pro
195 200 205
Ser Asn Thr Lys Val Asp Lys Lys Val Glu Pro Lys Ser Cys Asp Lys
210 215 220
Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro
225 230 235 240
Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser
245 250 255
Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp
260 265 270
Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn
275 280 285
Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val
290 295 300
Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu
305 310 315 320
Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys
325 330 335
Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr
340 345 350
Leu Pro Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr
355 360 365
Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu
370 375 380
Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu
385 390 395 400
Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys
405 410 415
Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu
420 425 430
Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly
435 440 445
Lys
<210> 10
<211> 446
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 10
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Tyr Met Tyr Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Gly Ile Asn Pro Ser Asn Gly Gly Thr Asn Phe Asn Glu Lys Phe
50 55 60
Lys Asn Arg Ala Thr Leu Thr Val Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Thr Arg Glu Gly Asn Pro Tyr Tyr Thr Met Asn Tyr Trp Gly Gln Gly
100 105 110
Thr Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe
115 120 125
Pro Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu
130 135 140
Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp
145 150 155 160
Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu
165 170 175
Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser
180 185 190
Ser Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His Lys Pro
195 200 205
Ser Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly Pro Pro
210 215 220
Cys Pro Pro Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser Val Phe
225 230 235 240
Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro
245 250 255
Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu Val
260 265 270
Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr
275 280 285
Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val Ser Val
290 295 300
Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys
305 310 315 320
Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser
325 330 335
Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro
340 345 350
Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val
355 360 365
Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly
370 375 380
Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp
385 390 395 400
Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg Trp
405 410 415
Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His
420 425 430
Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
435 440 445
<210> 11
<211> 446
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 11
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Tyr Met Tyr Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Gly Ile Asn Pro Ser Asn Ala Gly Thr Asn Phe Asn Glu Lys Phe
50 55 60
Lys Asn Arg Ala Thr Leu Thr Val Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Thr Arg Glu Gly Asn Pro Tyr Tyr Thr Met Asn Tyr Trp Gly Gln Gly
100 105 110
Thr Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe
115 120 125
Pro Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu
130 135 140
Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp
145 150 155 160
Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu
165 170 175
Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser
180 185 190
Ser Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His Lys Pro
195 200 205
Ser Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly Pro Pro
210 215 220
Cys Pro Pro Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser Val Phe
225 230 235 240
Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro
245 250 255
Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu Val
260 265 270
Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr
275 280 285
Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val Ser Val
290 295 300
Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys
305 310 315 320
Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser
325 330 335
Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro
340 345 350
Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val
355 360 365
Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly
370 375 380
Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp
385 390 395 400
Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg Trp
405 410 415
Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His
420 425 430
Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
435 440 445
<210> 12
<211> 449
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 12
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Tyr Met Tyr Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Gly Ile Asn Pro Ser Asn Gly Gly Thr Asn Phe Asn Glu Lys Phe
50 55 60
Lys Asn Arg Ala Thr Leu Thr Val Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Thr Arg Glu Gly Asn Pro Tyr Tyr Thr Met Asn Tyr Trp Gly Gln Gly
100 105 110
Thr Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe
115 120 125
Pro Leu Ala Pro Ser Ser Lys Ser Thr Ser Gly Gly Thr Ala Ala Leu
130 135 140
Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp
145 150 155 160
Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu
165 170 175
Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser
180 185 190
Ser Ser Leu Gly Thr Gln Thr Tyr Ile Cys Asn Val Asn His Lys Pro
195 200 205
Ser Asn Thr Lys Val Asp Lys Lys Val Glu Pro Lys Ser Cys Asp Lys
210 215 220
Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Ala Ala Gly Gly Pro
225 230 235 240
Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser
245 250 255
Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp
260 265 270
Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn
275 280 285
Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val
290 295 300
Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu
305 310 315 320
Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys
325 330 335
Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr
340 345 350
Leu Pro Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr
355 360 365
Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu
370 375 380
Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu
385 390 395 400
Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys
405 410 415
Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu
420 425 430
Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly
435 440 445
Lys
<210> 13
<211> 449
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 13
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Tyr Met Tyr Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Gly Ile Asn Pro Ser Asn Ala Gly Thr Asn Phe Asn Glu Lys Phe
50 55 60
Lys Asn Arg Ala Thr Leu Thr Val Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Thr Arg Glu Gly Asn Pro Tyr Tyr Thr Met Asn Tyr Trp Gly Gln Gly
100 105 110
Thr Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe
115 120 125
Pro Leu Ala Pro Ser Ser Lys Ser Thr Ser Gly Gly Thr Ala Ala Leu
130 135 140
Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp
145 150 155 160
Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu
165 170 175
Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser
180 185 190
Ser Ser Leu Gly Thr Gln Thr Tyr Ile Cys Asn Val Asn His Lys Pro
195 200 205
Ser Asn Thr Lys Val Asp Lys Lys Val Glu Pro Lys Ser Cys Asp Lys
210 215 220
Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Ala Ala Gly Gly Pro
225 230 235 240
Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser
245 250 255
Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp
260 265 270
Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn
275 280 285
Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val
290 295 300
Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu
305 310 315 320
Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys
325 330 335
Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr
340 345 350
Leu Pro Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr
355 360 365
Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu
370 375 380
Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu
385 390 395 400
Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys
405 410 415
Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu
420 425 430
Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly
435 440 445
Lys
<210> 14
<211> 446
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 14
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Tyr Met Tyr Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Gly Ile Asn Pro Ser Asn Gly Gly Thr Asn Phe Asn Glu Lys Phe
50 55 60
Lys Asn Arg Ala Thr Leu Thr Val Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Thr Arg Glu Gly Asn Pro Tyr Tyr Thr Met Asn Tyr Trp Gly Gln Gly
100 105 110
Thr Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe
115 120 125
Pro Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu
130 135 140
Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp
145 150 155 160
Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu
165 170 175
Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser
180 185 190
Ser Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His Lys Pro
195 200 205
Ser Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly Pro Pro
210 215 220
Cys Pro Pro Cys Pro Ala Pro Glu Phe Glu Gly Gly Pro Ser Val Phe
225 230 235 240
Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro
245 250 255
Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu Val
260 265 270
Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr
275 280 285
Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val Ser Val
290 295 300
Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys
305 310 315 320
Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser
325 330 335
Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro
340 345 350
Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val
355 360 365
Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly
370 375 380
Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp
385 390 395 400
Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg Trp
405 410 415
Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His
420 425 430
Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
435 440 445
<210> 15
<211> 446
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 15
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Tyr Met Tyr Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Gly Ile Asn Pro Ser Asn Ala Gly Thr Asn Phe Asn Glu Lys Phe
50 55 60
Lys Asn Arg Ala Thr Leu Thr Val Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Thr Arg Glu Gly Asn Pro Tyr Tyr Thr Met Asn Tyr Trp Gly Gln Gly
100 105 110
Thr Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe
115 120 125
Pro Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu
130 135 140
Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp
145 150 155 160
Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu
165 170 175
Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser
180 185 190
Ser Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His Lys Pro
195 200 205
Ser Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly Pro Pro
210 215 220
Cys Pro Pro Cys Pro Ala Pro Glu Phe Glu Gly Gly Pro Ser Val Phe
225 230 235 240
Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro
245 250 255
Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu Val
260 265 270
Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr
275 280 285
Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val Ser Val
290 295 300
Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys
305 310 315 320
Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser
325 330 335
Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro
340 345 350
Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val
355 360 365
Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly
370 375 380
Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp
385 390 395 400
Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg Trp
405 410 415
Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His
420 425 430
Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
435 440 445
<210> 16
<211> 446
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 16
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Tyr Met Tyr Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Gly Ile Asn Pro Ser Asn Gly Gly Thr Asn Phe Asn Glu Lys Phe
50 55 60
Lys Asn Arg Ala Thr Leu Thr Val Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Thr Arg Glu Gly Asn Pro Tyr Tyr Thr Met Asn Tyr Trp Gly Gln Gly
100 105 110
Thr Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe
115 120 125
Pro Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu
130 135 140
Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp
145 150 155 160
Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu
165 170 175
Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser
180 185 190
Ser Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His Lys Pro
195 200 205
Ser Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly Pro Pro
210 215 220
Cys Pro Pro Cys Pro Ala Pro Glu Ala Ala Gly Gly Pro Ser Val Phe
225 230 235 240
Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro
245 250 255
Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu Val
260 265 270
Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr
275 280 285
Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val Ser Val
290 295 300
Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys
305 310 315 320
Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser
325 330 335
Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro
340 345 350
Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val
355 360 365
Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly
370 375 380
Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp
385 390 395 400
Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg Trp
405 410 415
Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His
420 425 430
Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
435 440 445
<210> 17
<211> 446
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 17
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Tyr Met Tyr Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Gly Ile Asn Pro Ser Asn Ala Gly Thr Asn Phe Asn Glu Lys Phe
50 55 60
Lys Asn Arg Ala Thr Leu Thr Val Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Thr Arg Glu Gly Asn Pro Tyr Tyr Thr Met Asn Tyr Trp Gly Gln Gly
100 105 110
Thr Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe
115 120 125
Pro Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu
130 135 140
Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp
145 150 155 160
Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu
165 170 175
Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser
180 185 190
Ser Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His Lys Pro
195 200 205
Ser Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly Pro Pro
210 215 220
Cys Pro Pro Cys Pro Ala Pro Glu Ala Ala Gly Gly Pro Ser Val Phe
225 230 235 240
Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro
245 250 255
Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu Val
260 265 270
Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr
275 280 285
Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val Ser Val
290 295 300
Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys
305 310 315 320
Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser
325 330 335
Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro
340 345 350
Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val
355 360 365
Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly
370 375 380
Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp
385 390 395 400
Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg Trp
405 410 415
Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His
420 425 430
Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
435 440 445
<210> 18
<211> 219
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 18
Asp Val Val Met Thr Gln Ser Pro Leu Ser Leu Pro Val Thr Ile Gly
1 5 10 15
Gln Pro Ala Ser Ile Ser Cys Lys Ser Ser Gln Ser Leu Leu Glu Ser
20 25 30
Asp Gly Lys Thr Tyr Leu Asn Trp Leu Gln Gln Arg Pro Gly Gln Ser
35 40 45
Pro Arg Arg Leu Ile Tyr Leu Val Ser Lys Leu Asp Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Trp Gln Gly
85 90 95
Thr His Phe Pro Trp Thr Phe Gly Gly Gly Thr Lys Val Glu Ile Lys
100 105 110
Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu
115 120 125
Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe
130 135 140
Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln
145 150 155 160
Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser
165 170 175
Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu
180 185 190
Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser
195 200 205
Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys
210 215
<210> 19
<211> 10
<212> PRT
<213> Intelligent (Homo sapiens)
<400> 19
Phe Leu Ser Arg Pro Thr Glu Lys Thr Ile
1 5 10
<210> 20
<211> 10
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 20
Phe Trp Ser Arg Pro Thr Glu Lys Thr Ile
1 5 10
<210> 21
<211> 10
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 21
Phe Leu Ser Arg Pro Thr Glu Lys Thr Cys
1 5 10
<210> 22
<211> 10
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 22
Phe Leu Gly Arg Pro Thr Glu Lys Thr Ile
1 5 10
<210> 23
<211> 10
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 23
Phe Leu Ser Arg Pro Thr Glu Lys Asp Ile
1 5 10
<210> 24
<211> 10
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 24
Phe Leu Ser Arg Pro Thr Glu Lys Tyr Ile
1 5 10
<210> 25
<211> 10
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 25
Phe Leu Ser Arg Trp Thr Glu Lys Thr Ile
1 5 10
<210> 26
<211> 10
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 26
Phe Leu Ser Arg Pro Ser Glu Lys Thr Ile
1 5 10
<210> 27
<211> 10
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 27
Phe Leu Asn Arg Pro Thr Glu Lys Thr Ile
1 5 10
<210> 28
<211> 10
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 28
Phe Leu Ser Arg Pro Phe Glu Lys Thr Ile
1 5 10
<210> 29
<211> 10
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 29
Phe Leu Ser Arg Pro Thr Glu Lys Thr Gly
1 5 10
<210> 30
<211> 5
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 30
Phe Leu Ser Arg Pro
1 5
<210> 31
<211> 5
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 31
Leu Ser Arg Pro Thr
1 5
<210> 32
<211> 5
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 32
Ser Arg Pro Thr Glu
1 5
<210> 33
<211> 5
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 33
Arg Pro Thr Glu Lys
1 5
<210> 34
<211> 5
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 34
Pro Thr Glu Lys Thr
1 5
<210> 35
<211> 5
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 35
Thr Glu Lys Thr Ile
1 5
<210> 36
<211> 6
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 36
Phe Leu Ser Arg Pro Thr
1 5
<210> 37
<211> 6
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 37
Leu Ser Arg Pro Thr Glu
1 5
<210> 38
<211> 6
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 38
Ser Arg Pro Thr Glu Lys
1 5
<210> 39
<211> 6
<212> PRT
<213> unknown
<220>
<223> synthetic
<400> 39
Arg Pro Thr Glu Lys Thr
1 5

Claims (15)

1. An anti-Cx 43 antibody or antigen-binding fragment thereof, comprising:
first, second and third triple Complementary Determining Region (CDR) sequences having SEQ ID NOs 1, 2 and 3, respectively; and
first, second and third light chain CDR sequences having the amino acid sequences of SEQ ID NOS 4,5 and 6, respectively.
2. The antibody or fragment thereof of claim 1, comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO 7 and a light chain variable region having the amino acid sequence of SEQ ID NO 8.
3. An anti-Cx 43 antibody or antigen-binding fragment thereof, comprising a heavy chain having an amino acid sequence selected from SEQ ID NOS 9-17 and a light chain having an amino acid sequence of SEQ ID NO 18.
4. An anti-Cx 43 antibody or antigen-binding fragment thereof, which binds to an epitope within the amino acid sequence of FLSRPTEKTI (SEQ ID NO:19) when bound to Cx 43.
5. The antibody or fragment thereof of claim 4, wherein the epitope comprises one or more amino acid sequences selected from the group consisting of: f1, S3, R4, P5, T6, E7, K8, T9 and I10 of SEQ ID NO 19.
6. The antibody or fragment thereof of claim 4, wherein the epitope consists of F1, S3, R4, P5, T6, E7, K8, T9, and I10 of SEQ ID NO 19.
7. The antibody or fragment thereof of claim 4, wherein the epitope comprises SEQ ID NO:19, all ten amino acids.
8. The antibody or fragment thereof of claim 4, wherein the epitope consists of SEQ ID NO:19 of all ten amino acids.
9. An isolated anti-Cx 43 antibody or antigen binding fragment thereof, wherein the antibody or fragment thereof cross-competes for binding to Cx43 with the antibody or fragment thereof of any of claims 1-8.
10. An isolated anti-Cx 43 antibody or antigen binding fragment thereof, wherein said antibody or fragment thereof cross-competes for binding to Cx43 with the antibody or fragment thereof of any one of claims 1 to 3, wherein preferably said antibody or fragment thereof binds to an epitope located within the amino acid sequence of FLSRPTEKTI (SEQ ID NO:19), wherein more preferably the epitope comprises one or more of the amino acids SEQ ID NO:19 and I10, wherein even more preferably the epitope consists of SEQ ID NO:19 of all ten amino acids.
11. The antibody or fragment thereof of any one of claims 1-10, which promotes opening of a Cx43 hemichannel in bone cells.
12. A pharmaceutical composition for promoting Gx43 hemichannel opening in bone cells, preferably for the treatment of cancer, cancer metastasis, osteosarcoma, osteoporosis, or osteopenia, comprising the antibody or fragment thereof of any one of claims 1-11 and a pharmaceutically acceptable carrier.
13. Use of an antibody or fragment thereof according to any one of claims 1 to 11 in the manufacture of a medicament for promoting opening of a Cx43 hemichannel in bone cells, preferably for the treatment of cancer, cancer metastasis, osteoporosis or osteopenia.
14. A method of promoting opening of Cx43 hemichannels in bone cells, preferably for use in the treatment of cancer, cancer metastasis, osteosarcoma, osteoporosis or osteopenia, comprising contacting bone cells with an effective amount of an antibody or fragment thereof according to any one of claims 1 to 11.
15. A method for the treatment of a disease or disorder associated with Cx43 hemichannel opening in bone cells, preferably for the treatment of cancer, cancer metastasis, osteosarcoma, osteoporosis or osteopenia, comprising administering a therapeutically effective amount of an antibody or fragment thereof of any one of claims 1 to 11 to a patient in need thereof.
CN201980024875.6A 2018-04-02 2019-04-02 Connexin 43 antibodies and uses thereof Pending CN112368299A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310053523.1A CN116854816A (en) 2018-04-02 2019-04-02 Connexin 43 antibodies and uses thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201862651668P 2018-04-02 2018-04-02
US62/651,668 2018-04-02
PCT/US2019/025363 WO2019195273A1 (en) 2018-04-02 2019-04-02 Connexin 43 antibodies and use thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CN202310053523.1A Division CN116854816A (en) 2018-04-02 2019-04-02 Connexin 43 antibodies and uses thereof

Publications (1)

Publication Number Publication Date
CN112368299A true CN112368299A (en) 2021-02-12

Family

ID=68101187

Family Applications (2)

Application Number Title Priority Date Filing Date
CN201980024875.6A Pending CN112368299A (en) 2018-04-02 2019-04-02 Connexin 43 antibodies and uses thereof
CN202310053523.1A Pending CN116854816A (en) 2018-04-02 2019-04-02 Connexin 43 antibodies and uses thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN202310053523.1A Pending CN116854816A (en) 2018-04-02 2019-04-02 Connexin 43 antibodies and uses thereof

Country Status (6)

Country Link
US (1) US20210163581A1 (en)
JP (2) JP7437317B2 (en)
CN (2) CN112368299A (en)
AU (1) AU2019249427A1 (en)
CA (1) CA3095897A1 (en)
WO (1) WO2019195273A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113307871A (en) * 2020-02-27 2021-08-27 福州拓新天成生物科技有限公司 Preparation and application of novel anti-CD 19 antibody and CD19-CAR-T cell

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20220411494A1 (en) * 2019-02-04 2022-12-29 Alamab Therapeutics, Inc. Connexin 43 antibodies and use thereof
WO2020176844A1 (en) * 2019-02-28 2020-09-03 Board Of Regents, The University Of Texas System Compositions for treating osteosarcoma and methods of use
AU2020358101A1 (en) * 2019-10-02 2022-04-28 Alamab Therapeutics, Inc. Anto-connexin antibody formulations
CN116234576A (en) * 2020-07-31 2023-06-06 阿拉玛布治疗学股份有限公司 Anti-connexin antibody formulations

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017025016A1 (en) * 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
WO2017147561A1 (en) * 2016-02-26 2017-08-31 The Board Of Regents Of The University Of Texas System CONNEXIN (Cx) 43 HEMICHANNEL-BINDING ANTIBODIES AND USES THEREOF

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2723973A1 (en) * 2008-05-15 2009-11-19 Biogen Idec Ma Inc. Anti-fn14 antibodies and uses thereof
CA2921652A1 (en) * 2013-08-21 2015-02-26 Manuel A. Riquelme Compositions and methods for targeting connexin hemichannels
US9493552B2 (en) * 2013-11-15 2016-11-15 China Synthetic Rubber Corporation Therapeutic biologic for treatment of hepatocellular carcinoma

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017025016A1 (en) * 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
WO2017147561A1 (en) * 2016-02-26 2017-08-31 The Board Of Regents Of The University Of Texas System CONNEXIN (Cx) 43 HEMICHANNEL-BINDING ANTIBODIES AND USES THEREOF

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CHELIUS等: "Identification and characterization of deamidation sites in the conserved regions of human immunoglobulin gamma antibodies", 《ANAL.CHEM.》 *
CHELIUS等: "Identification and characterization of deamidation sites in the conserved regions of human immunoglobulin gamma antibodies", 《ANAL.CHEM.》, vol. 77, 31 December 2005 (2005-12-31) *
MANNING 等: "Stability of protein pharmaceuticals: An update", 《PHARMACEUTICAL RESEARCH》 *
MANNING 等: "Stability of protein pharmaceuticals: An update", 《PHARMACEUTICAL RESEARCH》, vol. 27, no. 4, 30 April 2010 (2010-04-30), pages 544 - 575, XP019793935, DOI: 10.1007/s11095-009-0045-6 *
WILLIAM R.STROHL AND LILA M.STROHL: "《Therapeutic antibody engineering》", 27 March 2014, WOODHEAD PUBLISHING SERIES IN BIOMEDICINE, pages: 378 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113307871A (en) * 2020-02-27 2021-08-27 福州拓新天成生物科技有限公司 Preparation and application of novel anti-CD 19 antibody and CD19-CAR-T cell

Also Published As

Publication number Publication date
CA3095897A1 (en) 2019-10-10
EP3774891A1 (en) 2021-02-17
CN116854816A (en) 2023-10-10
AU2019249427A1 (en) 2020-10-29
JP2021520393A (en) 2021-08-19
WO2019195273A1 (en) 2019-10-10
JP7437317B2 (en) 2024-02-22
US20210163581A1 (en) 2021-06-03
JP2024042116A (en) 2024-03-27

Similar Documents

Publication Publication Date Title
JP7437317B2 (en) Connexin 43 antibody and its use
ES2871112T3 (en) Neutralization of inhibitory pathways in lymphocytes
JP5908972B2 (en) Novel antigen binding protein
US20200147211A1 (en) Combination therapies for treating cancers
KR102428255B1 (en) Compositions and methods for detection and treatment of gastric cancer
CN107613974A (en) Anti- MET antibody and its application method
KR102497171B1 (en) Anti-angiopoietin-2 Antibodies and Uses Thereof
JP2024009998A (en) Anti-tissue factor antibody-drug conjugates and their use in treatment of cancer
JP2024026138A (en) How to treat cancer using a combination of platinum-based drugs and anti-tissue factor antibody-drug conjugates
KR102428254B1 (en) Compositions and methods for detection and treatment of ovarian cancer
KR20220054847A (en) AMHRII-binding antibody drug conjugates and their use in the treatment of cancer
US20220411494A1 (en) Connexin 43 antibodies and use thereof
TW201716439A (en) HER3 antibodies
KR20210086671A (en) Methods of treating cancer using a combination of an anti-VEGF antibody and an anti-tissue factor antibody-drug conjugate
JP2020515594A (en) ERBB-2 targeting agent comprising an antigen binding site that binds to an epitope on the extracellular portion of ERB-2 and ERBB-3 for the treatment of individuals with ERBB-2, ERBB-2/ERBB-3 positive tumors. And bispecific antibody
CN112040970A (en) Compositions and methods for cancer therapy

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40042791

Country of ref document: HK

RJ01 Rejection of invention patent application after publication
RJ01 Rejection of invention patent application after publication

Application publication date: 20210212